You are on page 1of 382

Essentials

of
Clinical Pathology

G
R
dV
te
ni
-U

ir
s.
ns
9
ir9

ia
rs
h
ta

e
p .p
vi

tahir99 - UnitedVRG
vip.persianss.ir
G
R
dV
te
ni
-U

ir
s.
ns
9
ir9

ia
rs
h
ta

e
p .p
vi

tahir99 - UnitedVRG
vip.persianss.ir
Essentials
of
Clinical Pathology

G
R
dV
te
Shirish M Kawthalkar
Associate Professor
ni
Department of Pathology
Government Medical College
-U

ir
Nagpur, Maharashtra, India

s.
ns
9
ir9

ia
rs
h
ta

e
p .p
vi

JAYPEE BROTHERS MEDICAL PUBLISHERS (P) LTD


Nagpur • St Louis (USA) • Panama City (Panama) • London (UK) • New Delhi • Ahmedabad
Bengaluru • Chennai • Hyderabad • Kochi • Kolkata • Lucknow • Mumbai

tahir99 - UnitedVRG
vip.persianss.ir
G
Published by
Jitendar P Vij

R
Jaypee Brothers Medical Publishers (P) Ltd
Corporate Office

dV
4838/24 Ansari Road, Daryaganj, New Delhi - 110002, India,
Phone: +91-11-43574357, Fax: +91-11-43574314
Registered Office
B-3 EMCA House, 23/23B Ansari Road, Daryaganj, New Delhi - 110 002, India
Phones: +91-11-23272143, +91-11-23272703, +91-11-23282021 te
ni
+91-11-23245672, Rel: +91-11-32558559, Fax: +91-11-23276490, +91-11-23245683
e-mail: jaypee@jaypeebrothers.com, Website: www.jaypeebrothers.com
-U

ir
Offices in India


Ahmedabad, Phone: Rel: +91-79-32988717, e-mail: ahmedabad@jaypeebrothers.com
Bengaluru, Phone: Rel: +91-80-32714073, e-mail: bangalore@jaypeebrothers.com
s.
ns
• Chennai, Phone: Rel: +91-44-32972089, e-mail: chennai@jaypeebrothers.com
9

• Hyderabad, Phone: Rel:+91-40-32940929, e-mail: hyderabad@jaypeebrothers.com


ir9

• Kochi, Phone: +91-484-2395740, e-mail: kochi@jaypeebrothers.com


ia

• Kolkata, Phone: +91-33-22276415, e-mail: kolkata@jaypeebrothers.com


• Lucknow, Phone: +91-522-3040554, e-mail: lucknow@jaypeebrothers.com
rs
h

• Mumbai, Phone: Rel: +91-22-32926896, e-mail: mumbai@jaypeebrothers.com


• Nagpur, Phone: Rel: +91-712-3245220, e-mail: nagpur@jaypeebrothers.com
ta

Overseas Offices
.p

• North America Office, USA, Ph: 001-636-6279734, e-mail: jaypee@jaypeebrothers.com, anjulav@jaypeebrothers.com


• Central America Office, Panama City, Panama, Ph: 001-507-317-0160, e-mail: cservice@jphmedical.com, Website: www.jphmedical.com
p

• Europe Office, UK, Ph: +44 (0) 2031708910, e-mail: info@jpmedpub.com


vi

Essentials of Clinical Pathology


© 2010, Shirish M Kawthalkar

All rights reserved. No part of this publication should be reproduced, stored in a retrieval system, or transmitted in any form or by any means:
electronic, mechanical, photocopying, recording, or otherwise, without the prior written permission of the author and the publisher.

This book has been published in good faith that the material provided by author is original. Every effort is made to ensure accuracy of material,
but the publisher, printer and author will not be held responsible for any inadvertent error (s). In case of any dispute, all legal matters are to be
settled under Delhi jurisdiction only.

First Edition: 2010


ISBN 978-93-80704-19-7
Typeset at JPBMP typesetting unit
Printed at

tahir99 - UnitedVRG
vip.persianss.ir
Preface

The major aims of this book are discussion of (i) use of laboratory tests in the investigation and management of
common diseases, and (ii) basic biochemical and pathological principles underlying the application of laboratory
tests. The book has been written keeping in mind mainly the curricula of undergraduate students of pathology. It
should also prove to be appropriate for postgraduate residents and students of medical laboratory technology. The
laboratory tests that are demonstrated to and performed by medical students in pathology practical class and during

G
university examination are given in more detail. To keep pace with new knowledge and advances, principles of
currently performed techniques in clinical laboratory practice have also been outlined. Most of the chapters are

R
followed by reference ranges and critical values for ready access. Critical values or action values are those laboratory
results that require immediate attention of the treating clinician. While interpreting results of laboratory tests, it is

dV
necessary to follow two fundamental rules of laboratory medicine: (i) diagnosis should never be made from a single
abnormal test result (since it is affected by a number of preanalytical and analytical factors), and (ii) try to arrive at
a single diagnosis (rather than multiple diagnoses) from all the abnormal test results obtained.

te
Clinical pathology is the second major subdivision of the discipline of pathology after anatomic pathology. It is
concerned with laboratory investigations for screening, diagnosis, and overall management of diseases by analysis
ni
of blood, urine, body fluids, and other specimens. The specialties included under the discipline of clinical pathology
are clinical chemistry, hematology, blood banking, medical microbiology, cytogenetics, and molecular genetics.
-U

ir
However, scope of this book does not allow microbiology and genetics to be included in this book.

s.
I must appreciate and recognize the unstinting support of my parents, my beloved wife Dr Anjali, and my two
children, Ameya and Ashish during preparation of this book. I am thankful to Dr HT Kanade, Dean, Government
ns
Medical College, Akola, Dr Smt Deepti Dongaonkar, Dean, Government Medical College, Nagpur, Dr BB Sonawane,
9

Professor and Head, Department of Pathology, Government Medical College, Akola, and Dr WK Raut, Professor
ir9

and Head, Department of Pathology, Government Medical College, Nagpur, for encouraging me in undertaking
ia

this project for the benefit of medical students.


I express my thanks to Mr JP Vij and his outstanding team of M/s Jaypee Brothers Medical Publishers for
rs
h

undertaking to publish this book, being patient with me during the preparation of the manuscript, and bringing it
ta

out in an easy-to-read and reader-friendly format.


e

Although I have made every effort to avoid any mistakes and errors, some may persist and feedback in this
.p

regard will be highly appreciated.


p

Shirish M Kawthalkar
vi

tahir99 - UnitedVRG
vip.persianss.ir
G
R
dV
te
ni
-U

ir
s.
ns
9
ir9

ia
rs
h
ta

e
p .p
vi

tahir99 - UnitedVRG
vip.persianss.ir
Contents

Section 1
Chemical Pathology and Related Studies

1. Examination of Urine ................................................................................................................................................. 3


2. Renal Function Tests ................................................................................................................................................ 30
3. Diabetes Mellitus ..................................................................................................................................................... 39
4. Liver Function Tests ................................................................................................................................................. 52
5. Disorders of Lipids and Biochemical Cardiac Markers .................................................................................... 69
6. Examination of Cerebrospinal Fluid .................................................................................................................... 80
7. Examination of Pleural and Peritoneal Fluids .................................................................................................... 91
8. Examination of Sputum........................................................................................................................................... 99
9. Examination of Feces ............................................................................................................................................. 104
10. Gastric Analysis ...................................................................................................................................................... 121
11. Tests for Malabsorption and Pancreatic Function ........................................................................................... 127
12. Thyroid Function Tests ......................................................................................................................................... 137
13. Pregnancy Tests ...................................................................................................................................................... 146
14. Infertility .................................................................................................................................................................. 150
15. Semen Analysis ....................................................................................................................................................... 159

Section 2
Laboratory Hematology
16. Hematopoiesis ......................................................................................................................................................... 169
17. Collection of Blood ................................................................................................................................................. 179
18. Estimation of Hemoglobin ................................................................................................................................... 183
19. Packed Cell Volume ............................................................................................................................................... 188
20. Total Leukocyte Count .......................................................................................................................................... 192
21. Reticulocyte Count ................................................................................................................................................. 196
22. Blood Smear ............................................................................................................................................................. 200
23. Red Cell Indices ...................................................................................................................................................... 213
24. Erythrocyte Sedimentation Rate .......................................................................................................................... 215
25. Examination of Bone Marrow .............................................................................................................................. 220
26. Diagnosis of Malaria and Other Parasites in Blood ........................................................................................ 229
27. Laboratory Tests in Anemia ................................................................................................................................. 244
28. Laboratory Tests in Hematological Malignancies ........................................................................................... 273
29. Laboratory Tests in Bleeding Disorders ............................................................................................................ 288
30. Laboratory Tests in Thrombophilia .................................................................................................................... 311
31. Laboratory Tests in Porphyrias ............................................................................................................................ 314
32. Automation in Hematology .................................................................................................................................. 319

tahir99 - UnitedVRG
vip.persianss.ir
viii Essentials of Clinical Pathology

Section 3
Practical Blood Transfusion

33. Blood Group Systems ............................................................................................................................................ 329


34. Blood Grouping ...................................................................................................................................................... 336
35. Collection of Donor Blood, Processing and Storage ........................................................................................ 341
36. Screening Tests for Infections Transmissible by Transfusion ...................................................................... 347
37. Compatibility Test (Cross-match) ....................................................................................................................... 352
38. Adverse Effects of Transfusion ............................................................................................................................ 354
39. Blood Components ................................................................................................................................................. 359

General References ...................................................................................................................................................... 365


Index ........................................................................................................................................................................... 367

tahir99 - UnitedVRG
vip.persianss.ir
Chemical Pathology and
Related Studies

tahir99 - UnitedVRG
vip.persianss.ir
tahir99 - UnitedVRG
vip.persianss.ir
1

Examination of Urine

COMPOSITION OF NORMAL URINE COLLECTION OF URINE


Urinalysis is one of the most commonly performed There are various methods for collection of urine. Method
laboratory tests in clinical practice. Composition of of collection to be used depends on the nature of
normal urine is shown in Table 1.1. investigation (Boxes 1.1 and 1.2).

Time of Collection
INDICATIONS FOR URINALYSIS
1. A single specimen: This may be a first morning
1. Suspected renal diseases like glomerulonephritis voiding, a random specimen, or a post-prandial
nephrotic syndrome, pyelonephritis, and renal failure specimen.
2. Detection of urinary tract infection The first voided specimen in the morning is the
most concentrated and has acidic pH in which formed
3. Detection and management of metabolic disorders
elements (cells and casts) are well preserved. This
like diabetes mellitus specimen is used for routine examination, fasting
4. Differential diagnosis of jaundice glucose, proteins, nitrite, microscopic analysis for
5. Detection and management of plasma cell dyscrasias cellular elements, pregnancy test, orthostatic
6. Diagnosis of pregnancy. proteinuria, and bacteriological analysis.

Table 1.1: Composition of normal urine (24 hour) in adults


Parameters Values

1. Volume 600-2000 ml
2. Specific gravity 1.003-1.030
3. Osmolality 300-900 mOsm/kg
4. pH 4.6-8.0
5. Glucose <0.5 gm
6. Proteins <150 mg
7. Urobilinogen 0.5-4.0 mg
8. Porphobilinogen 0-2 mg
9. Creatinine 14-26 mg/kg (men), 11-20 mg/kg (women)
10. Urea nitrogen 12-20 gm
11. Uric acid 250-750 mg
12. Sodium 40-220 mEq
13. Potassium 25-125 mEq
14. Chloride 110-250 mEq
15. Calcium (low calcium diet) 50-150 mg
16. Formiminoglutamic acid (FIGlu) < 3 mg
17. Red cells, epithelial cells, and white blood cells <1-2/high power field
4 Essentials of Clinical Pathology

Box 1.1: Collection of urine sample Box 1.2: Collection of urine for routine and culture
examination
• First morning, midstream: Preferred for routine urine Collection for routine urinalysis
examination. For routine examination of urine, a wide-mouthed glass bottle
• Random, midstream: Routine urine examination. of 20-30 ml capacity, which is dry, chemically clean, leak-
• First morning, midstream, clean catch: Bacteriological proof, and with a tight fitting stopper is used. About 15 ml
examination. of midstream sample is cleanly collected.
• Postprandial: Estimation of glucose, urobilinogen Collection for bacterial culture
• 24-hour: Quantitative estimation of proteins or hormones. • Use sterile container
• Catheterised: Bacteriological examination in infants, • Collect midstream, clean catch sample
bedridden patients, and in obstruction of urinary tract. • Must be plated within 2 hours of collection
• Plastic bag (e.g. colostomy bag) tied around genitals: • If refrigerated, must be plated within 24 hours of
Infants; incontinent adults. collection
• No preservative should be added.

The random specimen is a single specimen collected 3. Catheter specimen: This is used for bacteriological
at any time of day. It is used for routine urine exami- study or culture in bedridden, ill patients or in
nation. patients with obstruction of urinary tract. It is usually
Post-prandial specimen (collected 2 hours after a avoided in ambulatory patients since it carries the
meal in the afternoon) is sometimes requested for risk of introduction of infection.
estimation of glucose (to monitor insulin therapy in 4. Infants: In infants, a clean plastic bag can be attached
diabetes mellitus) or of urobilinogen. around the baby’s genitalia and left in place for some
2. 24-hour specimen: After getting up in the morning, time. For bacteriologic examination, urine is aspirated
the first urine is discarded. All the urine voided from bladder by passing a needle just above
subsequently during the rest of the day and the night symphysis pubis.
is collected in a large bottle (clean bottle of 2 liter
capacity with a cap). The first urine after getting up Changes which Occur in Standing Urine at
in the morning on the next day is also collected. The Room Temperature
urine should be preserved at 4-6°C during the period
of collection. The container is then immediately If urine is left standing at room temperature for long after
transported to the laboratory. The urine is thoroughly collection, following changes occur:
mixed and an aliquot is used for testing. This method • Increase in pH due to production of ammonia from
is used for quantitative estimation of proteins and urea by urease-producing bacteria.
hormones. • Formation of crystals due to precipitation of phos-
phates and calcium (making the urine turbid)
Collection Methods • Loss of ketone bodies, since they are volatile.
1. Midstream specimen: This is used for all types of • Decrease in glucose due to glycolysis and utilization
examinations. After voiding initial half of urine into of glucose by cells and bacteria.
the toilet, a part of urine is collected in the bottle. First • Oxidation of bilirubin to biliverdin causing false-
half of stream serves to flush out contaminating cells negative test for bilirubin
and microbes from urethra and perineum. Subse- • Oxidation of urobilinogen to urobilin causing false-
quent stream is collected which is from the urinary negative test for urobilinogen
bladder. • Bacterial proliferation
2. Clean-catch specimen: This is recommended for • Disintegration of cellular elements, especially in
bacteriologic culture. In men, glans penis is suffi- alkaline and hypotonic urine.
ciently exposed and cleaned with soap and water. In
women urethral opening should be exposed, washed
Urine sample must be tested in the laboratory within 2
with soapy cotton balls, rinsed with water-saturated
hours of collection to get the correct results.
cotton, and holding the labia apart, the initial urine
is allowed to pass into the toilet and the remaining is
Preservation of Urine Sample
voided into the bottle (amount 20-100 ml). This
method avoids contamination of urine with the The urine sample should ideally be examined within 1-2
vaginal fluids. hours of voiding. If delay in examination is expected,
Examination of Urine 5

then to slow down the above changes, sample can be Box 1.3: Physical examination
kept in the refrigerator for a maximum of 8 hours.
Refrigeration (4-6°C) is the best general method of • Volume • Odor
preservation up to 8 hours. Before analysis, refrigerated • Color • Specific gravity
samples should be warmed to room temperature. For • Appearance • pH
routine urinalysis, preservatives should be avoided, as
they interfere with reagent strip techniques and
chemical test for protein. Following chemical preser-
vatives can be added to the 24-hour urine sample: diabetes insipidus (failure of secretion of antidiuretic
• Hydrochloric acid: It is used for preservation of a 24- hormone), chronic renal failure (loss of concentrating
hour urine sample for adrenaline, noradrenaline, ability of kidneys) or diuretic therapy.
vanillylmandelic acid, and steroids. • Oliguria means urinary volume < 400 ml/24 hours.
• Toluene: It forms a thin layer over the surface and Causes include febrile states, acute glomerulo-
acts as a physical barrier for bacteria and air. It is used nephritis (decreased glomerular filtration), congestive
for measurement of chemicals. cardiac failure or dehydration (decreased renal blood
• Boric acid: A general preservative. flow).
• Thymol: It inhibits bacteria and fungi.
• Anuria means urinary output < 100 ml/24 hours or
• Formalin: It is an excellent chemical for preservation
of formed elements. complete cessation of urine output. It occurs in acute
tubular necrosis (e.g. in shock, hemolytic transfusion
PHYSICAL EXAMINATION reaction), acute glomerulonephritis, and complete
The parameters to be examined on physical examination urinary tract obstruction.
of urine are shown in Box 1.3.
Color
Volume
Volume of only the 24-hr specimen of urine needs to be Normal urine color in a fresh state is pale yellow or amber
measured and reported. The average 24-hr urinary and is due to the presence of various pigments
output in adults is 600-2000 ml. The volume varies collectively called urochrome. Depending on the state
according to fluid intake, diet, and climate. Abnormalities of hydration urine may normally be colorless (over
of urinary volume are as follows: hydration) or dark yellow (dehydration). Some of the
• Polyuria means urinary volume > 2000 ml/24 hours. abnormal colors with associated conditions are listed in
This is seen in diabetes mellitus (osmotic diuresis), Table 1.2.

Table 1.2: Different colors of urine

Colors Conditions

Colorless Dilute urine (diabetes mellitus, diabetes insipidus, overhydration)


Red Hematuria, Hemoglobinuria, Porphyria, Myoglobinuria
Dark brown or black Alkaptonuria, Melanoma
Brown Hemoglobinuria
Yellow Concentrated urine
Yellow-green or Biliverdin
green
Deep yellow with Bilirubin
yellow foam
Orange or orange- Urobilinogen
brown Porphobilinogen
Milky-white Chyluria
Red or orange Porphyria
fluorescence with
UV light

Note: Many drugs cause changes in urine color; drug history should be obtained if there is abnormal coloration of urine
6 Essentials of Clinical Pathology

Appearance Causes of decrease in SG of urine are diabetes insipidus


(SG consistently between 1.002-1.003), chronic renal
Normal, freshly voided urine is clear in appearance.
failure (low and fixed SG at 1.010 due to loss of
Causes of cloudy or turbid urine are listed in Table 1.3.
concentrating ability of tubules) and compulsive water
Foamy urine occurs in the presence of excess proteins or
drinking.
bilirubin.
Methods for measuring SG are urinometer method,
refractometer method, and reagent strip method.
Odor
1. Urinometer method: This method is based on the
Freshly voided urine has a typical aromatic odor due to principle of buoyancy (i.e. the ability of a fluid to exert
volatile organic acids. After standing, urine develops an upward thrust on a body placed in it). Urinometer
ammoniacal odor (formation of ammonia occurs when (a hydrometer) is placed in a container filled with
urea is decomposed by bacteria). Some abnormal odors urine (Fig. 1.1A). When solute concentration is high,
with associated conditions are: upthrust of solution increases and urinometer is
• Fruity: Ketoacidosis, starvation pushed up (high SG). If solute concentration is low,
• Mousy or musty: Phenylketonuria urinometer sinks further into the urine (low SG).
• Fishy: Urinary tract infection with Proteus, tyrosinae- Accuracy of a urinometer needs to be checked with
mia. distilled water. In distilled water, urinometer should
• Ammoniacal: Urinary tract infection with Escherichia
coli, old standing urine.
• Foul: Urinary tract infection
• Sulfurous: Cystinuria.

Specific Gravity (SG)


This is also called as relative mass density. It depends on
amount of solutes in solution. It is basically a comparison
of density of urine against the density of distilled water
at a particular temperature. Specific gravity of distilled
water is 1.000. Normal SG of urine is 1.003 to 1.030 and
depends on the state of hydration. SG of normal urine is
mainly related to urea and sodium. SG increases as solute
concentration increases and decreases when temperature
rises (since volume expands with rise in temperature).
SG of urine is a measure of concentrating ability of
kidneys and is determined to get information about
this tubular function. SG, however, is affected by
proteinuria and glycosuria.
Causes of increase in SG of urine are diabetes mellitus
(glycosuria), nephrotic syndrome (proteinuria), fever, Fig. 1.1: (A) Urinometer method and (B) Reagent strip
and dehydration. method for measuring specific gravity of urine

Table 1.3: Causes of cloudy or turbid urine

Cause Appearance Diagnosis

1. Amorphous phosphates White and cloudy on standing in Disappear on addition of a drop of


alkaline urine dilute acetic acid
2. Amorphous urates Pink and cloudy in acid urine Dissolve on warming
3. Pus cells Varying grades of turbidity Microscopy
4. Bacteria Uniformly cloudy; do not settle at the bottom Microscopy, Nitrite test
following centrifugation
Examination of Urine 7

show SG of 1.000 at the temperature of calibration. If not, 7.0). On standing, urine becomes alkaline because of loss
then the difference needs to be adjusted in test readings of carbon dioxide and production of ammonia from urea.
taken subsequently. Therefore, for correct estimation of pH, fresh urine
The method is as follows: should be examined.
1. Fill a measuring cylinder with 50 ml of urine. There are various methods for determination of
2. Lower urinometer gently into the urine and let it float reaction of urine: litmus paper, pH indicator paper, pH
freely.
meter, and reagent strip tests.
3. Let urinometer settle; it should not touch the sides or
bottom of the cylinder. 1. Litmus paper test: A small strip of litmus paper is
4. Take the reading of SG on the scale (lowest point of dipped in urine and any color change is noted. If blue
meniscus) at the surface of the urine. litmus paper turns red, it indicates acid urine. If red
5. Take out the urinometer and immediately note the paper turns blue, it indicates alkaline urine (Fig. 1.2A).
temperature of urine with a thermometer. 2. pH indicator paper: Reagent area (which is impreg-
Correction for temperature: Density of urine increases at nated with bromothymol blue and methyl red) of
low temperature and decreases at higher temperature. indicator paper strip is dipped in urine sample and
This causes false reading of SG. Therefore, SG is corrected the color change is compared with the color guide
for difference between urine temperature and calibration provided. Approximate pH is obtained.
temperature. Check the temperature of calibration of the
3. pH meter: An electrode of pH meter is dipped in urine
urinometer To get the corrected SG, add 0.001 to the
reading for every 3°C that the urine temperature is above sample and pH is read off directly from the digital
the temperature of calibration. Similarly subtract 0.001 display. It is used if exact pH is required.
from the reading for every 3°C below the calibration 4. Reagent strip test: The test area (Fig. 1.2B) contains
temperature. polyionic polymer bound to H+; on reaction with
Correction for dilution: If quantity of urine is not sufficient cations in urine, H+ is released causing change in color
for measurement of SG, urine can be appropriately of the pH-sensitive dye.
diluted and the last two figures of SG are multiplied by Normal pH range is 4.6 to 8.0 (average 6.0 or slightly
the dilution factor. acidic). Urine pH depends on diet, acid base balance,
Correction for abnormal solute concentration: High SG in the water balance, and renal tubular function.
presence of glycosuria or proteinuria will not reflect true Acidic urine is found in ketosis (diabetes mellitus,
kidney function (concentrating ability). Therefore it is starvation, fever), urinary tract infection by Escherichia
necessary to nullify the effect of glucose or proteins. For coli, and high protein diet. Alkaline urine may result from
this, 0.003 is subtracted from temperature-corrected SG
for each 1 gm of protein/dl urine and 0.004 for every 1
gm of glucose/dl urine.
2. Refractometer method: SG can be precisely deter-
mined by a refractometer, which measures the
refractive index of the total soluble solids. Higher the
concentration of total dissolved solids, higher the
refractive index. Extent of refraction of a beam of light
passed through urine is a measure of solute concen-
tration, and thus of SG. The method is simple and
requires only 1-2 drops of urine. Result is read from
a scale or from digital display.
3. Reagent strip method: Reagent strip (Fig. 1.1B)
measures the concentration of ions in urine, which
correlates with SG. Depending on the ionic strength
of urine, a polyelectrolyte will ionize in proportion.
This causes a change in color of pH indicator
(bromothymol blue).

Reaction and pH
The pH is the scale for measuring acidity or alkalinity Fig. 1.2: Testing pH of urine with litmus paper (A) and
(acid if pH is < 7.0; alkaline if pH is > 7.0; neutral if pH is with reagent strip test (B)
8 Essentials of Clinical Pathology

urinary tract infection by bacteria that split urea to Box 1.5: Causes of proteinuria
ammonia (Proteus or Pseudomonas), severe vomiting,
vegetarian diet, old ammoniacal urine sample and • Glomerular proteinuria
chronic renal failure. • Tubular proteinuria
Determining pH of urine helps in identifying various • Overflow proteinuria
crystals in urine. Altering pH of urine may be useful in • Hemodynamic (functional) proteinuria
treatment of renal calculi (i.e. some stones form only in • Post-renal proteinuria
acid urine e.g. uric acid calculi; in such cases urine is
kept alkaline); urinary tract infection (urine should be
kept acid); and treatment with certain drugs (e.g. disease, there is increased excretion of lower molecular
streptomycin is effective in urinary tract infection if urine weight proteins like albumin and transferrin. When
is kept alkaline). In unexplained metabolic acidosis, glomeruli can retain larger molecular weight proteins
measurement of urine pH is helpful in diagnosing renal but allow passage of comparatively lower molecular
tubular acidosis; in renal tubular acidosis, urine pH is weight proteins, the proteinuria is called as selective.
consistently alkaline despite metabolic acidosis. With further glomerular damage, this selectivity is lost
and larger molecular weight proteins (γ globulins) are
CHEMICAL EXAMINATION also excreted along with albumin; this is called as
nonselective proteinuria.
The chemical examination is carried out for substances
Selective and nonselective proteinuria can be distin-
listed in Box 1.4.
guished by urine protein electrophoresis. In selective
proteinuria, albumin and transferrin bands are seen,
Box 1.4: Chemical examination of urine
while in nonselective type, the pattern resembles that of
• Proteins • Urobilinogen serum (Fig. 1.3).
• Glucose • Blood Causes of glomerular proteinuria are glomerular
• Ketones • Hemoglobin diseases that cause increased permeability of glomerular
• Bilirubin • Myoglobin basement membrane. The degree of glomerular proteinu-
• Bile salts • Nitrite or leukocyte esterase

Proteins
Normally, kidneys excrete scant amount of protein in
urine (up to 150 mg/24 hours). These proteins include
proteins from plasma (albumin) and proteins derived
from urinary tract (Tamm-Horsfall protein, secretory
IgA, and proteins from tubular epithelial cells, leucocytes,
and other desquamated cells); this amount of proteinuria
cannot be detected by routine tests. (Tamm-Horsfall
protein is a normal mucoprotein secreted by ascending
limb of the loop of Henle).
Proteinuria refers to protein excretion in urine
greater than 150 mg/24 hours in adults.

Causes of Proteinuria
Causes of proteinuria can be grouped as shown in Box
1.5.
1. Glomerular proteinuria: Proteinuria due to increased
Fig. 1.3: Glomerular and tubular proteinuria. Upper figure shows
permeability of glomerular capillary wall is called as normal serum protein electrophoresis pattern. Lower part shows
glomerular proteinuria. comparison of serum and urine electrophoresis in (1) selective
There are two types of glomerular proteinuria: proteinuria, (2) non-selective proteinuria, and (3) tubular
selective and nonselective. In early stages of glomerular proteinuria
Examination of Urine 9

Box 1.6: Nephrotic syndrome and is probably due to lordotic posture that causes
inferior venacaval compression between the liver and
• Massive proteinuria (>3.5 gm/24 hr) vertebral column. The condition disappears in adulthood.
• Hypoalbuminemia (<3.0 gm/dl) Amount of proteinuria is <1000 mg/day. First-morning
• Generalised edema urine after rising is negative for proteins, while another
• Hyperlipidemia (serum cholesterol >350 mg/dl) urine sample collected after patient performs normal
• Lipiduria
activities is positive for proteins. In such patients, periodic
testing for proteinuria should be done to rule out renal
disease.
ria correlates with severity of disease and prognosis.
5. Post-renal proteinuria: This is caused by inflamma-
Serial estimations of urinary protein are also helpful in
monitoring response to treatment. Most severe degree tory or neoplastic conditions in renal pelvis, ureter,
of proteinuria occurs in nephrotic syndrome (Box 1.6). bladder, prostate, or urethra.
2. Tubular proteinuria: Normally, glomerular mem-
brane, although impermeable to high molecular Tests for Detection of Proteinuria
weight proteins, allows ready passage to low 1. Heat and acetic acid test (Boiling test): This test is
molecular weight proteins like β2-microglobulin, based on the principle that proteins get precipitated
retinol-binding protein, lysozyme, α1-microglobulin,
when boiled in an acidic solution.
and free immunoglobulin light chains. These low
molecular weight proteins are actively reabsorbed by Method: Urine should be clear; if not, filter or use
proximal renal tubules. In diseases involving mainly supernatant from a centrifuged sample.
tubules, these proteins are excreted in urine while Urine should be just acidic (check with litmus paper);
albumin excretion is minimal. if not, add 10% acetic acid drop by drop until blue litmus
Urine electrophoresis shows prominent α- and β- paper turns red.
bands (where low molecular weight proteins migrate) A test tube is filled 2/3rds with urine. The tube is
and a faint albumin band (Fig. 1.3). inclined at an angle and the upper portion is boiled over
Tubular type of proteinuria is commonly seen in
the flame. (Only the upper portion is heated so that
acute and chronic pyelonephritis, heavy metal
poisoning, tuberculosis of kidney, interstitial convection currents generated by heat do not disturb the
nephritis, cystinosis, Fanconi syndrome and rejection precipitate and the upper portion can be compared with
of kidney transplant. the lower clear portion). Compare the heated part with
Purely tubular proteinuria cannot be detected by the lower part. Cloudiness or turbidity indicates presence
reagent strip test (which is sensitive to albumin), but of either phosphates or proteins (Fig. 1.4). A few drops
heat and acetic acid test and sulphosalicylic acid test of 10% acetic acid are added and the upper portion is
are positive. boiled again. Turbidity due to phosphates disappears
3. Overflow proteinuria: When concentration of a low while that due to proteins does not.
molecular weight protein rises in plasma, it “over-
flows” from plasma into the urine. Such proteins are
immunoglobulin light chains or Bence Jones proteins
(plasma cell dyscrasias), hemoglobin (intravascular
hemolysis), myoglobin (skeletal muscle trauma), and
lysozyme (acute myeloid leukemia type M4 or M5).
4. Hemodynamic proteinuria: Alteration of blood flow
through the glomeruli causes increased filtration of
proteins. Protein excretion, however, is transient. It
is seen in high fever, hypertension, heavy exercise,
congestive cardiac failure, seizures, and exposure to
cold.
Postural (orthostatic) proteinuria occurs when the
subject is standing or ambulatory, but is absent in
recumbent position. It is common in adolescents (3-5%) Fig. 1.4: Principle of heat test for proteins
10 Essentials of Clinical Pathology

False-positive test occurs with tolbutamide and large


doses of penicillins.
2. Reagent strip test: The reagent area of the strip is
coated with an indicator and buffered to an acid pH
which changes color in the presence of proteins
(Figs 1.5 and 1.6). The principle is known as “protein
error of indicators”.
The reagent area is impregnated with bromo-
phenol blue indicator buffered to pH 3.0 with citrate.
When the dye gets adsorbed to protein, there is
change in ionization (and hence pH) of the indicator
that leads to change in color of the indicator. The
intensity of the color produced is proportional to the
concentration of protein. The test is semi-quantitative.
Reagent strip test is mainly reactive to albumin.
Fig. 1.6: Grading of proteinuria with reagent strip test
It is false-negative in the presence of Bence Jones (above) and sulphosalicylic acid test (below)
proteins, myoglobin, and hemoglobin. Overload
(Bence Jones) proteinuria and tubular proteinuria
denatured by organic acids and precipitate out of
may be missed entirely if only reagent strip method
solution).
is used. This test should be followed by sulpho-
Take 2 ml of clear urine in a test tube. If reaction of
salicylic acid test, which is a confirmatory test. Highly
urine is neutral or alkaline, a drop of glacial acetic acid is
alkaline urine, gross hematuria, and contamination
added. Add 2-3 drops of sulphosalicylic acid (3 to 5%),
with vaginal secretions can give false-positive
and examine for turbidity against a dark background
reactions. (Fig. 1.6).
3. Sulphosalicylic acid test: Addition of sulphosalicylic This test is more sensitive and reliable than boiling
acid to the urine causes formation of a white test.
precipitate if proteins are present (Proteins are False-positive test may occur due to gross hematuria,
highly concentrated urine, radiographic contrast media,
excess uric acid, tolbutamide, sulphonamides, salicylates,
and penicillins.
False-negative test can occur with very dilute urine.
The test can detect albumin, hemoglobin, myoglobin,
and Bence Jones proteins.
Comparison of reagent strip test and sulphosalicylic
acid test is shown in Table 1.4.
Fig. 1.5: Principle of reagent strip test for proteins. The principle
Quantitative Estimation of Proteins
is called as ‘protein error of indicators’ meaning that one color
appears if protein is present and another color if protein is Indications for quantitative estimation of proteins in
absent. Sensitivity is 5-10 mg/dl. The test does not detect Bence urine are:
Jones proteins, hemoglobin, and myoglobin • Diagnosis of nephrotic syndrome

Table 1.4: Comparison of two tests for proteinuria


Parameter Reagent strip test Sulphosalicylic acid test
1. Principle Colorimetric Acid precipitation
2. Proteins detected Albumin All (albumin, Bence Jones proteins,
hemoglobin, myoglobin)
3. Sensitivity 5-10 mg/dl 20 mg/dl
4. Indicator Color change Turbidity
5. Type of test Screening Confirmatory
Examination of Urine 11

Table 1.5: Grading of albuminuria


Condition mg/24 hr mg/L mg/g creatinine μg/min μg/mg creatinine g/mol creatinine
Normal < 30 < 20 < 20 < 20 < 30 < 2.5
Microalbuminuria 30-300 20-200 20-300 20-200 30-300 2.5-25
Overt albuminuria >300 >200 >300 >200 >300 >25

• Detection of microalbuminuria or early diabetic permeability to albumin and denotes microvascular


nephropathy disease. Microalbuminuria precedes the development
• To follow response to therapy in renal disease of diabetic nephropathy by a few years. If blood
Proteinuria >1500 mg/ 24 hours indicates glomerular glucose level and hypertension are tightly controlled
disease; proteinuria >3500 mg/24 hours is called as at this stage by aggressive treatment then progression
nephrotic range proteinuria; in tubular, hemodynamic to irreversible renal disease and subsequent renal
and post renal diseases, proteinuria is usually < 1500 mg/ failure can be delayed or prevented.
24 hours. 2. Microalbuminuria is an independent risk factor for
Grading of albuminuria is shown in Table 1.5. cardiovascular disease in diabetes mellitus.
There are two methods for quantitation of proteins:
(1) Estimation of proteins in a 24-hour urine sample, and Detection of microalbuminuria: Microalbuminuria cannot
be detected by routine tests for proteinuria. Methods for
(2) Estimation of protein/creatinine ratio in a random
detection include:
urine sample.
• Measurement of albumin-creatinine ratio in a random
1. Quantitative estimation of proteins in a 24-hour urine sample
urine sample: Collection of a 24-hour sample is given • Measurement of albumin in an early morning or
earlier. Adequacy of sample is confirmed by random urine sample
calculating expected 24-hour urine creatinine • Measurement of albumin in a 24 hr sample
excretion. Daily urinary creatinine excretion depends Test strips that screen for microalbuminuria are
on muscle mass and remains relatively constant in available commercially. Exact quantitation can be done
an individual patient. In adult males creatinine by immunologic assays like radioimmunoassay or
excretion is 14-26 mg/kg/24 hours, while in women enzyme linked immunosorbent assay.
it is 11-20 mg/kg/24 hours. Various methods are
available for quantitative estimation of proteins: Bence Jones Proteinuria
Esbach’s albuminometer method, turbidimetric
methods, biuret reaction, and immunologic methods. Bence Jones proteins are monoclonal immunoglobulin
2. Estimation of protein/creatinine ratio in a random light chains (either κ or λ) that are synthesized by
urine sample: Because of the problem of incomplete neoplastic plasma cells. Excess production of these light
collection of a 24-hour urine sample, many labora- chains occurs in plasma cell dyscrasias like multiple
tories measure protein/creatinine ratio in a random myeloma and primary amyloidosis. Because of their low
urine sample. Normal protein/creatinine ratio is molecular weight and high concentration they are
< 0.2. In low-grade proteinuria it is 0.2-1.0; in excreted in urine (overflow proteinuria).
moderate, it is 1.0-3.5; and in nephrotic- range Bence Jones proteins have a characteristic thermal
proteinuria it is > 3.5. behaviour. When heated, Bence Jones proteins precipi-
tate at temperatures between 40°C to 60°C (other proteins
Microalbuminuria precipitate between 60-70°C), and precipitate disappears
on further heating at 85-100°C (while precipitate of other
This is defined as urinary excretion of 30 to 300 mg/24
proteins does not). When cooled (60-85°C), there is
hours (or 2-20 mg/dl) of albumin in urine.
reappearance of precipitate of Bence Jones proteins. This
Significance of microalbuminuria test, however, is not specific for Bence Jones proteins and
1. Microalbuminuria is considered as the earliest sign both false-positive and -negative results can occur. This
of renal damage in diabetes mellitus (diabetic test has been replaced by protein electrophoresis of
nephropathy). It indicates increase in capillary concentrated urine sample (Fig. 1.7).
12 Essentials of Clinical Pathology

glucosuria or glycosuria (Box 1.7). Glycosuria results if


the filtered glucose load exceeds the capacity of renal
tubular reabsorption. Most common cause is hyper-
glycemia from diabetes mellitus.

Causes of Glycosuria
1. Glycosuria with hyperglycemia:
Fig. 1.7: Urine protein electrophoresis showing heavy Bence • Endocrine diseases: diabetes mellitus, acromegaly,
Jones proteinuria (red arrow) along with loss of albumin and Cushing’s syndrome, hyperthyroidism, pancrea-
other low molecular weight proteins in urine tic disease
• Non-endocrine diseases: central nervous system
diseases, liver disorders
Further evaluation of persistent overt proteinuria is • Drugs: adrenocorticotrophic hormone, cortico-
shown in Figure 1.8. steroids, thiazides
• Alimentary glycosuria (Lag-storage glycosuria):
Glucose After a meal, there is rapid intestinal absorption
of glucose leading to transient elevation of blood
The main indication for testing for glucose in urine is glucose above renal threshold. This can occur in
detection of unsuspected diabetes mellitus or follow-up persons with gastrectomy or gastrojejunostomy
of known diabetic patients. and in hyperthyroidism. Glucose tolerance test
Practically all of the glucose filtered by the glomeruli reveals a peak at 1 hour above renal threshold
is reabsorbed by the proximal renal tubules and returned (which causes glycosuria); the fasting and 2-hour
to circulation. Normally a very small amount of glucose glucose values are normal.
is excreted in urine (< 500 mg/24 hours or <15 mg/dl) 2. Glycosuria without hyperglycemia
that cannot be detected by the routine tests. Presence of • Renal glycosuria: This accounts for 5% of cases of
detectable amounts of glucose in urine is called as glycosuria in general population. Renal threshold

Note: Quantitation of proteins and creatinine clearance are done in all patients with persistent proteinuria

Fig. 1.8: Evaluation of proteinuria


Examination of Urine 13

Box 1.7: Urine glucose

• Urine should be tested for glucose within 2 hours of collection (due to lowering of glucose by glycolysis and by contaminating
bacteria which degrade glucose rapidly)
• Reagent strip test is a rapid, inexpensive, and semi-quantitative test
• In the past this test was used for home-monitoring of glucose; the test is replaced by glucometers.
• Urine glucose cannot be used to monitor control of diabetes since renal threshold is variable amongst individuals, no
information about level of blood glucose below renal threshold is obtained, and urinary glucose value is affected by
concentration of urine.

is the highest glucose level in blood at which Other carbohydrates (like lactose, fructose, galactose,
glucose appears in urine and which is detectable pentoses), certain metabolites (glucuronic acid, homo-
by routine laboratory tests. The normal renal gentisic acid, uric acid, creatinine), and drugs (ascorbic
threshold for glucose is 180 mg/dl. Threshold acid, salicylates, cephalosporins, penicillins, strepto-
substances need a carrier to transport them from mycin, isoniazid, para-aminosalicylic acid, nalidixic acid,
tubular lumen to blood. When the carrier is etc.) also reduce alkaline copper sulphate solution.
saturated, the threshold is reached and the
substance is excreted. Up to this level glucose Method
filtered by the glomeruli is efficiently reabsorbed 1. Take 5 ml of Benedict’s qualitative reagent in a test
by tubules. Renal glycosuria is a benign condition tube (composition of Benedict’s qualitative reagent:
in which renal threshold is set below 180 mgs/dl copper sulphate 17.3 gram, sodium carbonate 100
but glucose tolerance is normal; the disorder is gram, sodium citrate 173 gram, distilled water 1000
transmitted as autosomal dominant. Other ml).
conditions in which glycosuria can occur with 2. Add 0.5 ml (or 8 drops) of urine. Mix well.
blood glucose level remaining below 180 mgs/dl 3. Boil over a flame for 2 minutes.
are renal tubular diseases in which there is 4. Allow to cool at room temperature.
decreased glucose reabsorption like Fanconi’s 5. Note the color change, if any.
syndrome, and toxic renal tubular damage. During Sensitivity of the test is about 200 mg reducing
pregnancy, renal threshold for glucose is substance per dl of urine. Since Benedict’s test gives
decreased. Therefore it is necessary to estimate positive reaction with carbohydrates other than glucose,
blood glucose when glucose is first detected in it is also used as a screening test (for detection of
urine. galactose, lactose, fructose, maltose, and pentoses in
urine) for inborn errors of carbohydrate metabolism in
Tests for Detection of Glucose in Urine infants and children. For testing urine only for glucose,
reagent strips are preferred (see below).
1. Copper reduction methods The result is reported in grades as follows (Fig. 1.10):
A. Benedict’s qualitative test: When urine is boiled in Nil: no change from blue color
Benedict’s qualitative solution, blue alkaline copper Trace: Green without precipitate
sulphate is reduced to red-brown cuprous oxide if a 1+ (approx. 0.5 grams/dl): Green with precipitate
reducing agent is present (Fig. 1.9). The extent of 2+ (approx. 1.0 grams/dl): Brown precipitate
reduction depends on the concentration of the reducing 3+ (approx. 1.5 grams/dl: Yellow-orange precipitate
substance. This test, however, is not specific for glucose. 4+ (> 2.0 grams/dl): Brick- red precipitate.

Fig. 1.9: Principle of Benedict’s qualitative test for sugar in urine. Sensitivity is 200 mg of glucose/dl
14 Essentials of Clinical Pathology

Sensitivity of the test is about 100 mg glucose/dl of


urine.
False positive test occurs in the presence of oxidizing
agent (bleach or hypochlorite used to clean urine
containers), which oxidizes the chromogen directly.
False-negative test occurs in the presence of large
amounts of ketones, salicylates, ascorbic acid, and severe
Escherichia coli infection (catalase produced by organisms
in urine inactivates hydrogen peroxide).

Ketones
Excretion of ketone bodies (acetoacetic acid, β-hydroxy-
butyric acid, and acetone) in urine is called as ketonuria.
Fig. 1.10: Grading of Benedict’s test (above) and reagent Ketones are breakdown products of fatty acids and their
strip test (below) for glucose presence in urine is indicative of excessive fatty acid
metabolism to provide energy.

B. Clinitest tablet method (Copper reduction tablet test): This Causes of Ketonuria
is a modified form of Benedict’s test in which the reagents Normally ketone bodies are not detectable in the urine
are present in a tablet form (copper sulphate, citric acid,
of healthy persons. If energy requirements cannot be met
sodium carbonate, and anhydrous sodium hydroxide).
by metabolism of glucose (due to defective carbohydrate
Sensitivity is 200 mgs/dl of glucose.
metabolism, low carbohydrate intake, or increased
2. Reagent strip method This test is specific for glucose metabolic needs), then energy is derived from break-
and is therefore preferred over Benedict’s and Clinitest down of fats. This leads to the formation of ketone bodies
methods. It is based on glucose oxidase-peroxidase (Fig. 1.12).
reaction. Reagent area of the strips is impregnated with
two enzymes (glucose oxidase and peroxidase) and a 1. Decreased utilization of carbohydrates
chromogen. Glucose is oxidized by glucose oxidase with a. Uncontrolled diabetes mellitus with ketoacidosis: In
the resultant formation of hydrogen peroxide and diabetes, because of poor glucose utilization, there is
gluconic acid. Oxidation of chromogen occurs in the compensatory increased lipolysis. This causes
presence of hydrogen peroxide and the enzyme peroxi- increase in the level of free fatty acids in plasma.
dase with resultant color change (Fig. 1.11). Nature of Degradation of free fatty acids in the liver leads to
chromogen and buffer system differ in different strips. the formation of acetoacetyl CoA which then forms
The strip is dipped into the urine sample and color is ketone bodies. Ketone bodies are strong acids and
observed after a specified time and compared with the produce H+ ions, which are neutralized by bicar-
color chart provided (Fig. 1.10). bonate ions; fall in bicarbonate (i.e. alkali) level
This test is more sensitive than Benedict’s qualitative produces ketoacidosis. Ketone bodies also increase
test and specific only for glucose. Other reducing agents the plasma osmolality and cause cellular dehydration.
give negative reaction. Children and young adults with type 1 diabetes are

Fig. 1.11: Principle of reagent strip test for glucose in urine. Each mole of glucose produces one mole of peroxide,
and each mole of peroxide reduces one mole of oxygen. Sensitivity is 100 mg glucose/100 ml
Examination of Urine 15

No method for detection of ketonuria reacts with all


the three ketone bodies. Rothera’s nitroprusside method
and methods based on it detect acetoacetic acid and
acetone (the test is 10-20 times more sensitive to
acetoacetic acid than acetone). Ferric chloride test detects
acetoacetic acid only. β-hydroxybutyric acid is not
detected by any of the screening tests.
Methods for detection of ketone bodies in urine are
Rothera’s test, Acetest tablet method, ferric chloride test,
Fig. 1.12: Formation of ketone bodies. A small part of and reagent strip test.
acetoacetate is spontaneously and irreversibly converted to 1. Rothera’s’ test (Classic nitroprusside reaction) Acetoacetic
acetone. Most is converted reversibly to β-hydroxybutyrate
acid or acetone reacts with nitroprusside in alkaline
solution to form a purple-colored complex (Fig. 1.13).
Rothera’s test is sensitive to 1-5 mg/dl of acetoacetate
especially prone to ketoacidosis during acute illness and to 10-25 mg/dl of acetone.
and stress. If glycosuria is present, then test for ketone
bodies must be done. If both glucose and ketone Method
bodies are present in urine, then it indicates presence
of diabetes mellitus with ketoacidosis (Box 1.8). 1. Take 5 ml of urine in a test tube and saturate it with
In some cases of diabetes, ketone bodies are increased ammonium sulphate.
in blood but do not appear in urine. 2. Add a small crystal of sodium nitroprusside. Mix
Presence of ketone bodies in urine may be a warning well.
of impending ketoacidotic coma. 3. Slowly run along the side of the test tube liquor
b. Glycogen storage disease (von Gierke’s disease) ammonia to form a layer.
2. Decreased availability of carbohydrates in the diet: 4. Immediate formation of a purple permanganate
a. Starvation colored ring at the junction of the two fluids indicates
b. Persistent vomiting in children a positive test (Fig. 1.14).
c. Weight reduction program (severe carbohydrate False-positive test can occur in the presence of L-dopa
restriction with normal fat intake) in urine and in phenylketonuria.
3. Increased metabolic needs:
a. Fever in children 2. Acetest tablet test This is Rothera’s test in the form of a
b. Severe thyrotoxicosis tablet. The Acetest tablet consists of sodium nitro-
c. Pregnancy prusside, glycine, and an alkaline buffer. A purple-
d. Protein calorie malnutrition lavender discoloration of the tablet indicates the presence
of acetoacetate or acetone (≥ 5 mg/dl). A rough estimate
Tests for Detection of Ketones in Urine of the amount of ketone bodies can be obtained by
The proportion of ketone bodies in urine in ketosis is comparison with the color chart provided by the
variable: β-hydroxybutyric acid 78%, acetoacetic acid manufacturer.The test is more sensitive than reagent strip
20%, and acetone 2%. test for ketones.

Box 1.8: Urine ketones in diabetes

Indications for testing


• At diagnosis of diabetes mellitus
• At regular intervals in all known cases of diabetes,
and in gestational diabetes
• In known diabetic patients during acute illness, persistent Fig. 1.13: Principles of Rothera’s test and reagent strip test
hyperglycemia (>300 mg/dl), pregnancy, clinical evidence for ketone bodies in urine. Ketones are detected as acetoacetic
of diabetic acidosis (nausea, vomiting, abdominal pain) acid and acetone but not β-hydroxybutyric acid
16 Essentials of Clinical Pathology

Table 1.6: Urine bilirubin and urobilinogen in jaundice

Urine test Hemolytic Hepatocellular Obstructive


jaundice jaundice jaundice

1. Bilirubin Absent Present Present


2. Urobilinogen Increased Increased Absent

In acute viral hepatitis, bilirubin appears in urine


even before jaundice is clinically apparent. In a fever
of unknown origin bilirubinuria suggests hepatitis.

Presence of bilirubin in urine indicates conjugated


hyperbilirubinemia (obstructive or hepatocellular
jaundice). This is because only conjugated bilirubin is
water-soluble. Bilirubin in urine is absent in hemolytic
jaundice; this is because unconjugated bilirubin is
Fig. 1.14: Rothera’s tube test and reagent strip test for
water-insoluble.
ketone bodies in urine

Tests for Detection of Bilirubin in Urine


3. Ferric chloride test (Gerhardt’s): Addition of 10% ferric
Bilirubin is converted to non-reactive biliverdin on
chloride solution to urine causes solution to become
exposure to light (daylight or fluorescent light) and on
reddish or purplish if acetoacetic acid is present. The test
standing at room temperature. Biliverdin cannot be
is not specific since certain drugs (salicylate and L-dopa)
detected by tests that detect bilirubin. Therefore fresh
give similar reaction. Sensitivity of the test is 25-50 mg/
dl. sample that is kept protected from light is required.
Findings associated with bilirubinuria are shown in
4. Reagent strip test: Reagent strips tests are modifications Box 1.9.
of nitroprusside test (Figs 1.13 and 1.14). Their sensitivity Methods for detection of bilirubin in urine are foam
is 5-10 mg/dl of acetoacetate. If exposed to moisture, test, Gmelin’s test, Lugol iodine test, Fouchet’s test,
reagent strips often give false-negative result. Ketone pad Ictotest tablet test, and reagent strip test.
on the strip test is especially vulnerable to improper 1. Foam test: About 5 ml of urine in a test tube is shaken
storage and easily gets damaged. and observed for development of yellowish foam.
Similar result is also obtained with proteins and
Bile Pigment (Bilirubin) highly concentrated urine. In normal urine, foam is
Bilirubin (a breakdown product of hemoglobin) is white.
undetectable in the urine of normal persons. Presence of 2. Gmelin’s test: Take 3 ml of concentrated nitric acid
bilirubin in urine is called as bilirubinuria. in a test tube and slowly place equal quantity of urine
There are two forms of bilirubin: conjugated and over it. The tube is shaken gently; play of colors
unconjugated. After its formation from hemoglobin in (yellow, red, violet, blue, and green) indicates positive
reticuloendothelial system, bilirubin circulates in blood test (Fig. 1.15).
bound to albumin. This is called as unconjugated 3. Lugol iodine test: Take 4 ml of Lugol iodine solution
bilirubin. Unconjugated bilirubin is not water-soluble, (Iodine 1 gm, potassium iodide 2 gm, and distilled
is bound to albumin, and cannot pass through the water to make 100 ml) in a test tube and add 4 drops
glomeruli; therefore it does not appear in urine. The liver of urine. Mix by shaking. Development of green color
takes up unconjugated bilirubin where it combines with indicates positive test.
glucuronic acid to form bilirubin diglucuronide
(conjugated bilirubiun). Conjugated bilirubin is water-
soluble, is filtered by the glomeruli, and therefore appears Box 1.9: Clinical and laboratory findings in bilirubinuria
in urine. • Jaundice
Detection of bilirubin in urine (along with urobili- • Urine color: Dark yellow with yellow foam
nogen) is helpful in the differential diagnosis of • Elevated serum conjugated bilirubin
jaundice (Table 1.6).
Examination of Urine 17

Fig. 1.16: Positive Fouchet’s test for bilirubin in urine

bile salts and conjugated bilirubin regurgitate into blood


from biliary canaliculi (due to increased intrabiliary
pressure) and are excreted in urine. The test used for their
Fig. 1.15: Positive Gmelin’s test for bilirubin showing detection is Hay’s surface tension test. The property of
play of colors bile salts to lower the surface tension is utilized in this
test.
4. Fouchet’s test: This is a simple and sensitive test. Take some fresh urine in a conical glass tube. Urine
i. Take 5 ml of fresh urine in a test tube, add 2.5 should be at the room temperature. Sprinkle on the
ml of 10% of barium chloride, and mix well. A surface particles of sulphur. If bile salts are present,
precipitate of sulphates appears to which bilirubin sulphur particles sink to the bottom because of lowering
is bound (barium sulphate-bilirubin complex). of surface tension by bile salts. If sulphur particles remain
ii. Filter to obtain the precipitate on a filter paper. on the surface of urine, bile salts are absent.
iii. To the precipitate on the filter paper, add 1drop Thymol (used as a preservative) gives false positive
of Fouchet’s reagent. (Fouchet’s reagent consists test.
of 25 grams of trichloroacetic acid, 10 ml of 10%
ferric chloride, and distilled water 100 ml). Urobilinogen
iv. Immediate development of blue-green color
Conjugated bilirubin excreted into the duodenum
around the drop indicates presence of bilirubin
through bile is converted by bacterial action to urobilino-
(Fig. 1.16).
5. Reagent strips or tablets impregnated with diazo gen in the intestine. Major part is eliminated in the feces.
reagent: These tests are based on reaction of bilirubin A portion of urobilinogen is absorbed in blood, which
with diazo reagent; color change is proportional to undergoes recycling (enterohepatic circulation); a small
the concentration of bilirubin. Tablets (Ictotest) detect amount, which is not taken up by the liver, is excreted in
0.05-0.1 mg of bilirubin/dl of urine; reagent strip tests urine. Urobilinogen is colorless; upon oxidation it is
are less sensitive (0.5 mg/dl). converted to urobilin, which is orange-yellow in color.
Normally about 0.5-4 mg of urobilinogen is excreted in
Bile Salts urine in 24 hours. Therefore, a small amount of urobili-
Bile salts are salts of four different types of bile acids: nogen is normally detectable in urine.
cholic, deoxycholic, chenodeoxycholic, and lithocholic. Urinary excretion of urobilinogen shows diurnal
These bile acids combine with glycine or taurine to form variation with highest levels in afternoon. Therefore, a
complex salts or acids. Bile salts enter the small intestine 2-hour post-meal sample is preferred.
through the bile and act as detergents to emulsify fat and
reduce the surface tension on fat droplets so that enzymes Causes of Increased Urobilinogen in Urine
(lipases) can breakdown the fat. In the terminal ileum,
bile salts are absorbed and enter in the blood stream from 1. Hemolysis: Excessive destruction of red cells leads
where they are taken up by the liver and re-excreted in to hyperbilirubinemia and therefore increased
bile (enterohepatic circulation). formation of urobilinogen in the gut. Bilirubin, being
Bile salts along with bilirubin can be detected in urine of unconjugated type, does not appear in urine.
in cases of obstructive jaundice. In obstructive jaundice, Increased urobilinogen in urine without bilirubin is
18 Essentials of Clinical Pathology

typical of hemolytic anemia. This also occurs in


megaloblastic anemia due to premature destruction
of erythroid precursors in bone marrow (ineffective
erythropoiesis).
2. Hemorrhage in tissues: There is increased formation
of bilirubin from destruction of red cells.

Causes of Reduced Urobilinogen in Urine


1. Obstructive jaundice: In biliary tract obstruction,
delivery of bilirubin to the intestine is restricted and
very little or no urobilinogen is formed. This causes
stools to become clay-colored.
2. Reduction of intestinal bacterial flora: This prevents
conversion of bilirubin to urobilinogen in the
intestine. It is observed in neonates and following
antibiotic treatment.
Testing of urine for both bilirubin and urobilinogen
can provide helpful information in a case of jaundice
(Table 1.6). Fig. 1.17: Ehrlich’s aldehyde test for urobilinogen

Tests for Detection of Urobilinogen in Urine


test is used. Add 1-2 ml of chloroform, shake for 2
Fresh urine sample should be used because on standing minutes and allow to stand. Pink color in the chloroform
urobilinogen is converted to urobilin, which cannot be layer indicates presence of urobilinogen, while pink
detected by routine tests. A timed (2-hour postprandial) coloration of aqueous portion indicates presence of
sample can also be used for testing urobilinogen. porphobilinogen. Pink layer is then decanted and shaken
Methods for detection of increased amounts of urobili- with butanol. A pink color in the aqueous layer indicates
nogen in urine are Ehrlich’s aldehyde test and reagent porphobilinogen (Fig. 1.18).
strip test. False-negative reaction can occur in the presence of
1. Ehrlich’s aldehyde test: Ehrlich’s reagent (p- (i) urinary tract infection (nitrites oxidize urobilinogen
dimethylaminobenzaldehyde) reacts with urobili- to urobilin), and (ii) antibiotic therapy (gut bacteria which
nogen in urine to produce a pink color. Intensity of produce urobilinogen are destroyed).
color developed depends on the amount of urobili- 2. Reagent strip method: This method is specific for
nogen present. Presence of bilirubin interferes with urobilinogen. Test area is impregnated with either
the reaction, and therefore if present, should be p-dimethylaminobenzaldehyde or 4-methoxy-
removed. For this, equal volumes of urine and 10% benzene diazonium tetrafluoroborate.
barium chloride are mixed and then filtered. Test for
urobilinogen is carried out on the filtrate. However, Blood
similar reaction is produced by porphobilinogen (a
The presence of abnormal number of intact red blood
substance excreted in urine in patients of porphyria).
cells in urine is called as hematuria. It implies presence
Method: Take 5 ml of fresh urine in a test tube. Add 0.5 of a bleeding lesion in the urinary tract. Bleeding in urine
ml of Ehrlich’s aldehyde reagent (which consists of may be noted macroscopically or with naked eye (gross
hydrochloric acid 20 ml, distilled water 80 ml, and para- hematuria). If bleeding is noted only by microscopic
dimethylaminobenzaldehyde 2 gm). Allow to stand at examination or by chemical tests, then it is called as
room temperature for 5 minutes. Development of pink occult, microscopic or hidden hematuria.
color indicates normal amount of urobilinogen. Dark
Causes of Hematuria
red color means increased amount of urobilinogen (Fig.
1.17). 1. Diseases of urinary tract
Since both urobilinogen and porphobilinogen • Glomerular diseases: Glomerulonephritis, Berger’s
produce similar reaction, further testing is required to disease, lupus nephritis, Henoch-Schonlein
distinguish between the two. For this, Watson-Schwartz purpura
Examination of Urine 19

Fig. 1.18: Interpretation of Watson-Schwartz test

• Nonglomerular diseases: Calculus, tumor, infec- cells) as well as myoglobin. Heme proteins in
tion, tuberculosis, pyelonephritis, hydronephrosis, hemoglobin act as peroxidase, which reduces
polycystic kidney disease, trauma, after strenuous hydrogen peroxide to water. This process needs a
physical exercise, diseases of prostate (benign hydrogen donor (benzidine, orthotoluidine, or
hyperplasia of prostate, carcinoma of prostate). guaiac). Oxidation of hydrogen donor leads to
2. Hematological conditions: Coagulation disorders, sickle development of a color (Fig. 1.19). Intensity of color
cell disease produced is proportional to the amount of hemo-
Presence of red cell casts and proteinuria along with
globin present.
hematuria suggests glomerular cause of hematuria.
Chemical tests are positive in hematuria, hemo-
Tests for Detection of Blood in Urine globinuria, and myoglobinuria.
1. Microscopic examination of urinary sediment: • Benzidine test: Make saturated solution of benzidine
Definition of microscopic hematuria is presence of 3 in glacial acetic acid. Mix 1 ml of this solution with 1
or more number of red blood cells per high power ml of hydrogen peroxide in a test tube. Add 2 ml of
field on microscopic examination of urinary sediment urine. If green or blue color develops within 5
in two out of three properly collected samples. A
minutes, the test is positive.
small number of red blood cells in urine of low specific
• Orthotoluidine test: In this test, instead of benzidine,
gravity may undergo lysis, and therefore hematuria
may be missed if only microscopic examination is orthotoluidine is used. It is more sensitive than
done. Therefore, microscopic examination of urine benzidine test.
should be combined with a chemical test. • Reagent strip test: Various reagent strips are
2. Chemical tests: These detect both intracellular and commercially available which use different
extracellular hemoglobin (i.e. intact and lysed red chromogens (o-toluidine, tetramethylbenzidine).

Fig. 1.19: Principle of chemical test for red cells, hemoglobin, or myoglobin in urine
20 Essentials of Clinical Pathology

Fig. 1.20: Evaluation of positive chemical test for blood in urine

Causes of false-positive tests: glucose-6-phosphate dehydrogenase deficiency


• Contamination of urine by menstrual blood in following exposure to oxidant drugs, immune
females hemolysis (mismatched blood transfusion, paroxy-
• Contamination of urine by oxidizing agent (e.g. smal cold hemoglobinuria), paroxysmal nocturnal
hypochlorite or bleach used to clean urine containers), hemoglobinuria, hemolytic uremic syndrome, and
or microbial peroxidase in urinary tract infection.
disseminated intravascular coagulation.
Causes of false-negative tests:
• Presence of a reducing agent like ascorbic acid in high Tests for Detection of Hemoglobinuria
concentration: Microscopic examination for red cells
is positive but chemical test is negative. Tests for detection of hemoglobinuria are benzidine test,
• Use of formalin as a preservative for urine orthotoluidine test, and reagent strip test.
Evaluation of positive chemical test for blood is
shown in Figure 1.20. Hemosiderin
Hemosiderin in urine (hemosiderinuria) indicates
Hemoglobin
presence of free hemoglobin in plasma. Hemosiderin
Presence of free hemoglobin in urine is called as appears as blue granules when urine sediment is stained
hemoglobinuria. with Prussian blue stain (Fig. 1.21). Granules are located
inside tubular epithelial cells or may be free if cells have
Causes of Hemoglobinuria
disintegrated. Hemosiderinuria is seen in intravascular
1. Hematuria with subsequent lysis of red blood cells hemolysis.
in urine of low specific gravity.
2. Intravascular hemolysis: Hemoglobin will appear in Myoglobin
urine when haptoglobin (to which hemoglobin binds
in plasma) is completely saturated with hemoglobin. Myoglobin is a protein present in striated muscle (skeletal
Intravascular hemolysis occurs in infections (severe and cardiac) which binds oxygen. Causes of myoglo-
falciparum malaria, clostridial infection, E. coli binuria include injury to skeletal or cardiac muscle, e.g.
septicemia), trauma to red cells (march hemo- crush injury, myocardial infarction, dermatomyositis,
globinuria, extensive burns, prosthetic heart valves), severe electric shock, and thermal burns.
Examination of Urine 21

reagent strip format that can detect significant


bacteriuria: nitrite test and leucocyte esterase test. These
tests are helpful at places where urine microscopy is not
available. If these tests are positive, urine culture is
indicated.
1. Nitrite test: Nitrites are not present in normal urine;
ingested nitrites are converted to nitrate and excreted
in urine. If gram-negative bacteria (e.g. E.coli,
Salmonella, Proteus, Klebsiella, etc.) are present in urine,
they will reduce the nitrates to nitrites through the
action of bacterial enzyme nitrate reductase. Nitrites
are then detected in urine by reagent strip tests. As E.
coli is the commonest organism causing urinary tract
infection, this test is helpful as a screening test for
urinary tract infection.
Some organisms like Staphylococci or Pseudomonas do
not reduce nitrate to nitrite and therefore in such
infections nitrite test is negative. Also, urine must be
retained in the bladder for minimum of 4 hours for
Fig. 1.21: Staining of urine sediment with Prussian blue
stain to demonstrate hemosiderin granules (blue)
conversion of nitrate to nitrite to occur; therefore, fresh
early morning specimen is preferred. Sufficient dietary
intake of nitrate is necessary. Therefore a negative nitrite
Chemical tests used for detection of blood or test does not necessarily indicate absence of urinary
hemoglobin also give positive reaction with myoglobin tract infection.
(as both hemoglobin and myoglobin have peroxidase The test detects about 70% cases of urinary tract
activity). Ammonium sulfate solubility test is used as a infections.
screening test for myoglobinuria (Myoglobin is soluble 2. Leucocyte esterase test: It detects esterase enzyme
in 80% saturated solution of ammonium sulfate, while released in urine from granules of leucocytes. Thus
hemoglobin is insoluble and is precipitated. A positive the test is positive in pyuria. If this test is positive,
chemical test for blood done on supernatant indicates urine culture should be done. The test is not sensitive
myoglobinuria). to leucocytes < 5/HPF.
Distinction between hematuria, hemoglobinuria, and
myoglobinuria is shown in Table 1.7. MICROSCOPIC EXAMINATION
Chemical Tests for Significant Bacteriuria Microscopic examination of urine is also called as the
(Indirect Tests for Urinary Tract Infection) “liquid biopsy of the urinary tract”.
In addition to direct microscopic examination of urine Urine consists of various microscopic, insoluble, solid
sample, chemical tests are commercially available in a elements in suspension. These elements are classified as

Table 1.7: Differentiation between hematuria, hemoglobinuria, and myoglobinuria


Parameter Hematuria Hemoglobinuria Myoglobinuria

1. Urine color Normal, smoky, red, Pink, red, or Red or brown


or brown brown
2. Plasma color Normal Pink Normal
3. Urine test based on Positive Positive Positive
peroxidase activity
4. Urine microscopy Many red cells Occasional red cell Occasional red cell
5. Serum haptoglobin Normal Low Normal
6. Serum creatine kinase Normal Normal Markedly increased
22 Essentials of Clinical Pathology

organized or unorganized. Organized substances examined within 2 hours of voiding because cells and
include red blood cells, white blood cells, epithelial cells, casts degenerate upon standing at room temperature. If
casts, bacteria, and parasites. The unorganized sub- preservative is required, then 1 crystal of thymol or 1
stances are crystalline and amorphous material. These drop of formalin (40%) is added to about 10 ml of urine.
elements are suspended in urine and on standing they
settle down and sediment at the bottom of the container; Method: A well-mixed sample of urine (12 ml) is
therefore they are known as urinary deposits or urinary centrifuged in a centrifuge tube for 5 minutes at 1500
sediments. Examination of urinary deposit is helpful in rpm and supernatant is poured off. The tube is tapped
diagnosis of urinary tract diseases as shown in Table 1.8. at the bottom to resuspend the sediment (in 0.5 ml of
Different types of urinary sediments are shown in urine). A drop of this is placed on a glass slide and
Figure 1.22. The major aim of microscopic examination covered with a cover slip (Fig. 1.23). The slide is examined
of urine is to identify different types of cellular elements immediately under the microscope using first the low
and casts. Most crystals have little clinical significance. power and then the high power objective. The condenser
Specimen: The cellular elements are best preserved in should be lowered to better visualize the elements by
acid, hypertonic urine; they deteriorate rapidly in reducing the illumination.
alkaline, hypotonic solution. A mid-stream, freshly
voided, first morning specimen is preferred since it is Cells
the most concentrated. The specimen should be Cellular elements in urine are shown in Figure 1.24.

Table 1.8: Urinary findings in renal diseases

Condition Albumin RBCs/HPF WBCs/HPF Casts/LPF Others

1. Normal 0-trace 0-2 0-2 Occasional –


(Hyaline)
2. Acute 1-2+ Numerous; 0-few Red cell, Smoky urine or
glomerulonephritis dysmorphic granular hematuria
3. Nephrotic syndrome >4+ 0-few 0-few Fatty, hyaline, Oval fat bodies,
Waxy, epithelial lipiduria
4. Acute pyelonephritis 0-1+ 0-few Numerous WBC, granular WBC clumps,
bacteria, nitrite test

HPF: High power field; LPF: Low power field; RBCs: Red blood cells; WBCs: White blood cells.

Fig. 1.22: Different types of urinary sediment

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Urine 23

Red Blood Cells

Normally there are no or an occasional red blood cell in


urine. In a fresh urine sample, red cells appear as small,
smooth, yellowish, anucleate biconcave disks about 7 μ
in diameter (called as isomorphic red cells). However,
red cells may appear swollen (thin discs of greater
diameter, 9-10 μ) in dilute or hypotonic urine, or may
appear crenated (smaller diameter with spikey surface)
in hypertonic urine. In glomerulonephritis, red cells are
typically described as being dysmorphic (i.e. markedly
variable in size and shape). They result from passage of
red cells through the damaged glomeruli. Presence of
> 80% of dysmorphic red cells is strongly suggestive of
glomerular pathology.
The quantity of red cells can be reported as number
Fig. 1.23: Preparation of urine sediment for of red cells per high power field.
microscopic examination Causes of hematuria have been listed earlier.

Fig. 1.24: Cells in urine (1) Isomorphic red blood cells, (2) Crenated red cells, (3) Swollen red cells, (4) Dysmorphic red
cells, (5) White blood cells (pus cells), (6) Squamous epithelial cell, (7) Transitional epithelial cells, (8) Renal tubular epithelial
cells, (9) Oval fat bodies, (10) Maltese cross pattern of oval fat bodies, and (11) spermatozoa

tahir99 - UnitedVRG
vip.persianss.ir
24 Essentials of Clinical Pathology

White Blood Cells (Pus Cells) diamond- or pear-shaped (caudate cells). Large numbers
or sheets of these cells in urine occur after catheterization
White blood cells are spherical, 10-15 μ in size, granular
and in transitional cell carcinoma.
in appearance in which nuclei may be visible. Degene-
rated white cells are distorted, smaller, and have fewer Oval Fat Bodies
granules. Clumps of numerous white cells are seen in These are degenerated renal tubular epithelial cells filled
infections. Presence of many white cells in urine is called with highly refractile lipid (cholesterol) droplets. Under
as pyuria. In hypotonic urine white cells are swollen and polarized light, they show a characteristic “Maltese
the granules are highly refractile and show Brownian cross” pattern. They can be stained with a fat stain such
movement; such cells are called as glitter cells; large as Sudan III or Oil Red O. They are seen in nephrotic
numbers are indicative of injury to urinary tract. syndrome in which there is lipiduria.
Normally 0-2 white cells may be seen per high power
field. Pus cells greater than 10/HPF or presence of Spermatozoa
clumps is suggestive of urinary tract infection. They may sometimes be seen in urine of men.
Increased numbers of white cells occur in fever,
pyelonephritis, lower urinary tract infection, tubulo- Telescoped urinary sediment: This refers to urinary
interstitial nephritis, and renal transplant rejection. sediment consisting of red blood cells, white blood cells,
In urinary tract infection, following are usually seen oval fat bodies, and all types of casts in roughly equal
in combination: proportion. It occurs in lupus nephritis, malignant
• Clumps of pus cells or pus cells >10/HPF hypertension, rapidly proliferative glomerulonephritis,
and diabetic glomerulosclerosis.
• Bacteria
• Albuminuria Organisms
• Positive nitrite test
Organisms detectable in urine are shown in Figure 1.25.
Simultaneous presence of white cells and white cell
casts indicates presence of renal infection (pyelo- Bacteria
nephritis). Bacteria in urine can be detected by microscopic
Eosinophils (>1% of urinary leucocytes) are a examination, reagent strip tests for significant bacteriuria
characteristic feature of acute interstitial nephritis due to (nitrite test, leucocyte esterase test), and culture.
drug reaction (better appreciated with a Wright’s stain). Method of collection for bacteriologic examination
is given earlier in Box 1.2.
Renal Tubular Epithelial Cells
Significant bacteriuria exists when there are >105
Presence of renal tubular epithelial cells is a significant bacterial colony forming units/ml of urine in a clean-
finding. Increased numbers are found in conditions catch midstream sample, >104 colony forming units/ml
causing tubular damage like acute tubular necrosis, of urine in catheterized sample, and >10 3 colony-
pyelonephritis, viral infection of kidney, allograft forming units/ml of urine in a suprapubic aspiration
rejection, and salicylate or heavy metal poisoning. sample.
These cells are small (about the same size or slightly
larger than white blood cell), polyhedral, columnar, or
oval, and have granular cytoplasm. A single, large,
refractile, eccentric nucleus is often seen.
Renal tubular epithelial cells are difficult to distin-
guish from pus cells in unstained preparations.

Squamous Epithelial Cells


Squamous epithelial cells line the lower urethra and
vagina. They are best seen under low power objective
(×10). Presence of large numbers of squamous cells in
urine indicates contamination of urine with vaginal fluid.
These are large cells, rectangular in shape, flat with
abundant cytoplasm and a small, central nucleus.

Transitional Epithelial Cells


Transitional cells line renal pelvis, ureters, urinary Fig. 1.25: Organisms in urine: (A) Bacteria, (B) Yeasts,
bladder, and upper urethra. These cells are large, and (C) Trichomonas, and (D) Egg of Schistosoma haematobium

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Urine 25

1. Microscopic examination: In a wet preparation, mellitus. Usually pyuria is present if there is infection
presence of bacteria should be reported only when by Candida. Candida may also be a contaminant in the
urine is fresh. Bacteria occur in combination with pus sample and therefore urine sample must be examined in
cells. Gram’s-stained smear of uncentrifuged urine a fresh state.
showing 1 or more bacteria per oil-immersion field
Trichomonas vaginalis
suggests presence of > 105 bacterial colony forming
units/ml of urine. If many squamous cells are present, These are motile organisms with pear shape, undulating
then urine is probably contaminated with vaginal membrane on one side, and four flagellae. They cause
flora. Also, presence of only bacteria without pus cells vaginitis in females and are thus contaminants in urine.
indicates contamination with vaginal or skin flora. They are easily detected in fresh urine due to their
2. Chemical or reagent strip tests for significant motility.
bacteriuria: These are given earlier.
3. Culture: On culture, a colony count of >105/ml is Eggs of Schistosoma haematobium
strongly suggestive of urinary tract infection, even Infection by this organism is prevalent in Egypt.
in asymptomatic females. Positive culture is followed
by sensitivity test. Most infections are due to Gram- Microfilariae
negative enteric bacteria, particularly Escherichia coli. They may be seen in urine in chyluria due to rupture of
If three or more species of bacteria are identified on a urogenital lymphatic vessel.
culture, it almost always indicates contamination by
vaginal flora. Casts
Negative culture in the presence of pyuria (‘sterile’ Urinary casts are cylindrical, cigar-shaped microscopic
pyuria) occurs with prior antibiotic therapy, renal structures that form in distal renal tubules and collecting
tuberculosis, prostatitis, renal calculi, catheterization, ducts. They take the shape and diameter of the lumina
fever in children (irrespective of cause), female genital (molds or ‘casts’) of the renal tubules. They have parallel
tract infection, and non-specific urethritis in males. sides and rounded ends. Their length and width may be
variable. Casts are basically composed of a precipitate of
Yeast Cells (Candida) a protein that is secreted by tubules (Tamm-Horsfall
These are round or oval structures of approximately the protein). Since casts form only in renal tubules their
same size as red blood cells. In contrast to red cells, they presence is indicative of disease of the renal parenchyma.
show budding, are oval and more refractile, and are not Although there are several types of casts, all urine casts
soluble in 2% acetic acid. are basically hyaline; various types of casts are formed
Presence of Candida in urine may suggest immuno- when different elements get deposited on the hyaline
compromised state, vaginal candidiasis, or diabetes material (Fig. 1.26). Casts are best seen under low power

Fig. 1.26: Genesis of casts in urine. All cellular casts degenerate to granular and waxy casts

tahir99 - UnitedVRG
vip.persianss.ir
26 Essentials of Clinical Pathology

objective (×10) with condenser lowered down to reduce muscle exercise in healthy persons and during fever.
the illumination. Increased numbers are found in conditions causing
Casts are the only elements in the urinary sediment glomerular proteinuria.
that are specifically of renal origin.
Granular casts: Presence of degenerated cellular debris in
Casts (Fig. 1.27) are of two main types:
a cast makes it granular in appearance. These are
• Noncellular: Hyaline, granular, waxy, fatty
cylindrical structures with coarse or fine granules (which
• Cellular: Red blood cell, white blood cell, renal represent degenerated renal tubular epithelial cells)
tubular epithelial cell. embedded in Tamm-Horsfall protein matrix. They are
Hyaline and granular casts may appear in normal or seen after strenuous muscle exercise and in fever, acute
diseased states. All other casts are found in kidney glomerulonephritis, and pyelonephritis.
diseases.
Waxy cast: These are the most easily recognized of all
Non-cellular Casts casts. They form when hyaline casts remain in renal
tubules for long time (prolonged stasis). They have
Hyaline casts: These are the most common type of casts
homogenous, smooth glassy appearance, cracked or
in urine and are homogenous, colorless, transparent, and
serrated margins and irregular broken-off ends. The ends
refractile. They are cylindrical with parallel sides and
are straight and sharp and not rounded as in other casts.
blunt, rounded ends and low refractive index. Presence
They are light yellow in color. They are most commonly
of occasional hyaline cast is considered as normal. Their
seen in end-stage renal failure.
presence in increased numbers (“cylinduria”) is
abnormal. They are composed primarily of Tamm- Fatty casts: These are cylindrical structures filled with
Horsfall protein. They occur transiently after strenuous highly refractile fat globules (triglycerides and cholesterol

Fig. 1.27: Urinary casts: (A) Hyaline cast, (B) Granular cast, (C) Waxy cast, (D) Fatty cast, (E) Red cell cast,
(F) White cell cast, and (G) Epithelial cast

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Urine 27

esters) in Tamm-Horsfall protein matrix. They are seen Normal Crystals


in nephrotic syndrome.
Crystals present in acid urine
Broad casts: Broad casts form in dilated distal tubules and a. Uric acid crystals: These are variable in shape
are seen in chronic renal failure and severe renal tubular (diamond, rosette, plates), and yellow or red-brown
obstruction. Both waxy and broad casts are associated in color (due to urinary pigment). They are soluble in
with poor prognosis. alkali, and insoluble in acid. Increased numbers are
found in gout and leukemia. Flat hexagonal uric acid
Cellular Casts crystals may be mistaken for cysteine crystals that also
form in acid urine.
To be called as cellular, casts should contain at least three b. Calcium oxalate crystals: These are colorless, refractile,
cells in the matrix. Cellular casts are named according to and envelope-shaped. Sometimes dumbbell-shaped
the type of cells entrapped in the matrix. or peanut-like forms are seen. They are soluble in
Red cell casts: These are cylindrical structures with red dilute hydrochloric acid. Ingestion of certain foods
cells in Tamm-Horsfall protein matrix. They may appear like tomatoes, spinach, cabbage, asparagus, and
brown in color due to hemoglobin pigmentation. These rhubarb causes increase in their numbers. Their
have greater diagnostic importance than any other cast. increased number in fresh urine (oxaluria) may also
If present, they help to differentiate hematuria due to suggest oxalate stones. A large number are seen in
glomerular disease from hematuria due to other causes. ethylene glycol poisoning.
RBC casts usually denote glomerular pathology e.g. acute c. Amorphous urates: These are urate salts of potassium,
glomerulonephritis. magnesium, or calcium in acid urine. They are usually
yellow, fine granules in compact masses. They are
White cell casts: These are cylindrical structures with white soluble in alkali or saline at 60°C.
blood cells embedded in Tamm-Horsfall protein matrix.
Leucocytes usually enter into tubules from the inter- Crystals present in alkaline urine:
stitium and therefore presence of leucocyte casts indicates a. Calcium carbonate crystals: These are small, colorless,
tubulointerstitial disease like pyelonephritis. and grouped in pairs. They are soluble in acetic acid
and give off bubbles of gas when they dissolve.
Renal tubular epithelial cell casts: These are composed of b. Phosphates: Phosphates may occur as crystals (triple
renal tubular epithelial cells that have been sloughed off. phosphates, calcium hydrogen phosphate), or as
They are seen in acute tubular necrosis, viral renal amorphous deposits.
disease, heavy metal poisoning, and acute allograft • Phosphate crystals
rejection. Even an occasional renal tubular cast is a  Triple phosphates (ammonium magnesium
significant finding.
phosphate): They are colorless, shiny, 3-6 sided
prisms with oblique surfaces at the ends (“coffin-
Crystals
lids”), or may have a feathery fern-like appearance.
Crystals are refractile structures with a definite geometric  Calcium hydrogen phosphate (stellar phosphate):
shape due to orderly 3-dimensional arrangement of its These are colorless, and of variable shape (star-
atoms and molecules. Amorphous material (or deposit) shaped, plates or prisms).
has no definite shape and is commonly seen in the form • Amorphous phosphates: These occur as colorless
of granular aggregates or clumps. small granules, often dispersed.
Crystals in urine (Fig. 1.28) can be divided into two All phosphates are soluble in dilute acetic acid.
main types: (1) Normal (seen in normal urinary
sediment), and (2) Abnormal (seen in diseased states). c. Ammonium urate crystals: These occur as cactus-like
However, crystals found in normal urine can also be seen (covered with spines) and called as ‘thornapple’
in some diseases in increased numbers. crystals. They are yellow-brown and soluble in acetic
Most crystals have no clinical importance acid at 60°C.
(particularly phosphates, urates, and oxalates). Crystals
can be identified in urine by their morphology. However, Abnormal Crystals
before reporting presence of any abnormal crystals, it is They are rare, but result from a pathological process.
necessary to confirm them by chemical tests. These occur in acid pH, often in large amounts. Abnormal

tahir99 - UnitedVRG
vip.persianss.ir
28 Essentials of Clinical Pathology

Fig. 1.28: Crystals in urine. (A) Normal crystals: (1) Calcium oxalate, (2) Triple phosphates, (3) Uric acid, (4) Amorphous
phosphates, (5) Amorphous urates, (6) Ammonium urate. (B) Abnormal crystals: (1) Cysteine, (2) Cholesterol, (3) Bilirubin,
(4) Tyrosine, (5) Sulfonamide, and (6) Leucine

crystals should not be reported on microscopy alone; and appear stacked in a stair-step arrangement. They
additional chemical tests are done for confirmation. are soluble in ether, chloroform, or alcohol. They are
1. Cysteine crystals: These are colorless, clear, hexagonal seen in lipiduria e.g. nephrotic syndrome and hyper-
(having 6 sides), very refractile plates in acid urine. cholesterolemia. They can be positively identified by
polarizing microscope.
They often occur in layers. They are soluble in 30%
3. Bilirubin crystals: These are small (5 μ), brown crystals
hydrochloric acid. They are seen in cysteinuria, an of variable shape (square, bead-like, or fine needles).
inborn error of metabolism. Cysteine crystals are often Their presence can be confirmed by doing reagent
associated with formation of cysteine stones. strip or chemical test for bilirubin. These crystals are
2. Cholesterol crystals: These are colorless, refractile, flat soluble in strong acid or alkali. They are seen in severe
rectangular plates with notched (missing) corners, obstructive liver disease.

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Urine 29

4. Leucine crystals: These are refractile, yellow or brown, CRITICAL FINDINGS


spheres with radial or concentric striations. They are
soluble in alkali. They are usually found in urine along • Strongly positive test for glucose and ketone bodies
with tyrosine in severe liver disease (cirrhosis). • Positive test for reducing sugar in an infant
5. Tyrosine crystals: They appear as clusters of fine, • Hemoglobinuria
delicate, colorless or yellow needles and are seen in • Red cell casts or >50% dysmorphic red cells on
liver disease and tyrosinemia (an inborn error of microscopic examination
metabolism). They dissolve in alkali. • Abnormal crystals like cysteine, leucine, or tyrosine.
6. Sulfonamide crystals: They are variably shaped
crystals, but usually appear as sheaves of needles.
BIBLIOGRAPHY
They occur following sulfonamide therapy. They are
soluble in acetone. 1. Burtis CA, Ashwood ER (Eds). Tietz fundamentals of
clinical chemistry (5th Ed). Philadelphia; WB Saunders
REFERENCE RANGES Company, 2001.
2. Carroll MF, Temte JL. Proteinuria in adults: A diagnostic
Volume in 24 hours: Adults: 600-2000 ml
approach. Am Fam Physician 2000;62:1333-40.
Color: Pale yellow to colorless
3. Cheesbrough M. District laboratory practice in tropical
Appearance: Clear countries. Part 1 and Part 2. Cambridge; Cambridge
Odor: Aromatic University Press, 1998.
Specific gravity: 1.003-1.030 4. Grossfeld GD, Wolf JS, Litwin MS, et al. Asymptomatic
Osmolality: 300-900 mOsm/kg of water microscopic hematuria in adults: Summary of the AUA
pH: 4.6-8.0 (Average: 6.0) best policy recommendations. Am Fam Physician 2001;
Proteins: Qualitative test: Negative 63:1145-54.
Quantitative test: < 150 mg/24 hours 5. Henry JB (Ed): Clinical diagnosis and management by
Albumin: < 30 mg/24 hours laboratory methods. (20th Ed). Philadelphia; WB Saunders
Glucose: Qualitative test: Negative Company, 2001.
Quantitative test: < 500 mg/24 hours (< 15 mg/dl) 6. King M. A medical laboratory for developing countries.
Ketones: Qualitative test: Negative London. Oxford University Press, 1973.
7. Mathieson PW. The cellular basis of albuminuria. Clinical
Bilirubin: Negative
Science 2004;107:533-8.
Bile salts: Negative
8. Simerville JA, Maxted WC, Pahira JJ. Urinalysis: A
Occult blood: Negative
comprehensive review. Am Fam Physician 2005;71:
Urobilinogen: 0.5-4.0 mg/24 hours 1153-62.
Myoglobin (Ammonium sulphate solubility test): 9. Wallach J. Interpretation of diagnostic tests. (7th Ed).
Negative Philadelphia. Lippincott Williams and Wilkins, 2000.
Microscopy: 1-2 red cells, pus cells, or epithelial cells/ 10. World Health Organization. Manual of basic techniques
HPF; occasional hyaline cast/LPF; Phosphate, oxalate, for a health laboratory (2nd Ed). Geneva; World Health
or urate crystals depending on urine pH. Organization, 2003.

tahir99 - UnitedVRG
vip.persianss.ir
2

Renal Function Tests

Kidney is a highly specialized organ that performs Box 2.1: Conditions with increased risk of
following functions: chronic renal disease
• Maintenance of extracellular fluid volume and
• Diabetes mellitus
composition: Kidney regulates water and electrolyte • Hypertension
balance, acid-base balance, and fluid osmotic • Autoimmune diseases like systemic lupus erythematosus
pressure. • Older age (GFR declines with age)
• Excretion of metabolic waste products (blood urea, • Family history of renal disease
creatinine, uric acid) and drugs, but retention of • Systemic infection
essential substances (like glucose and amino acids). • Urinary tract infection
• Lower urinary tract obstruction
• Regulation of blood pressure by renin-angiotensin
mechanism
• Synthesis of erythropoietin, a hormone which 4. Plan renal replacement therapy (dialysis or renal
stimulates erythropoiesis transplantation) in advanced renal disease.
• Production of vit. D3 (active form of vit. D) from vit. 5. Adjust dosage of certain drugs (e.g. chemotherapy)
D2, which stimulates absorption of calcium from according to renal function.
gastrointestinal tract.

FACTORS AFFECTING RENAL FUNCTION CLASSIFICATION OF


RENAL FUNCTION TESTS
Kidney function is affected by following factors:
• Diffuse renal disease. Renal function tests can be classified as shown in Table
• Pre-renal conditions—Decreased renal blood flow as 2.1.
in dehydration, congestive cardiac failure and shock. In practice, the commonly performed renal function
• Post-renal conditions—Obstruction to urinary tests are routine urinalysis, estimation of serum
outflow. creatinine, blood urea nitrogen (BUN), BUN/Serum
creatinine ratio, creatinine clearance test (or estimation
INDICATIONS FOR RENAL FUNCTION TESTS of GFR from serum creatinine value by a prediction
1. Early identification of impairment of renal function equation), and estimation of urine concentrating ability
in patients with increased risk of chronic renal (water deprivation test). Urine examination is the first
disease: Early detection and treatment of renal test performed in patients suspected of having renal
impairment in chronic renal disease prevent compli- disease. It is the simplest and the least expensive renal
cations of chronic renal failure and is associated with function test. In urine examination parameters that can
improved prognosis. Laboratory tests can be applied assess renal function are urine volume in 24 hours,
in individuals who are at increased risk of developing specific gravity, osmolality, proteinuria, and microscopic
chronic renal disease (Box 2.1) to detect renal examination of urinary sediment.
functional impairment at an early stage and to detect
degree of kidney damage. Tests to Evaluate Glomerular Function
2. Diagnosis of renal disease The best test to assess overall kidney function is
3. Follow the course of renal disease and assess estimation of glomerular filtration rate or GFR (Box 2.2).
response to treatment. GFR varies according to age, sex, and body surface area.

tahir99 - UnitedVRG
vip.persianss.ir
Renal Function Tests 31

Table 2.1: Classification of renal function tests

Tests to evaluate glomerular function Tests to evaluate tubular function

1. Clearance tests to measure glomerular 1. Tests to assess proximal tubular


filtration rate: Inulin clearance, 125I-iothalamate function:
clearance, 51Cr-EDTA clearance, Cystatin C • Glycosuria, phosphaturia, uricosuria
clearance, Creatinine clearance, and Urea • Generalized aminoaciduria
clearance • Tubular proteinuria
2. Calculation of creatinine clearance from • Fractional sodium excretion
prediction equations 2. Tests to assess distal tubular function:
3. Blood biochemistry: Serum creatinine, • Specific gravity and osmolality of urine
Blood urea nitrogen (BUN), and BUN/serum creatinine ratio • Water-deprivation test and water-loading test
4. Microalbuminuria and albuminuria • Ammonium chloride loading test

Box 2.2: Glomerular filtration rate (GFR) • Stage 3: Moderately reduced GFR (GFR 30-59 ml/
min/1.73 m2)
• Best test for assessment of excretory renal function • Stage 4: Severely reduced GFR (GFR 15-29 ml/min/
• Varies according to age, sex, and body weight of an 1.73 m2)
individual; a normal GFR also depends on normal
• Stage 5: Kidney failure (GFR < 15 ml/min/1.73 m2)
renal blood flow and pressure.
• Normal GFR in young adults is 120-130 ml/min per Kidney damage refers to presence of pathological
1.73 m2. abnormalities or markers of damage like abnormalities
• Creatinine clearance is commonly used as a measure in blood or urine tests or imaging studies. Symptoms
of GFR. Equations can be used to estimate GFR usually develop at or after stage 3. GFR <60 ml/min per
from serum creatinine value. 1.73 m2 indicates loss of ≥ 50% of kidney function. GFR
• GFR declines with age (due to glomerular arteriolo- <15 ml/min per 1.73 m2 is associated with kidney failure
sclerosis) and uremia. Following methods are used to measure
• GFR <60 ml/min per 1.73 m2 indicates loss of ≥50% GFR: (1) Clearance tests and (2) Prediction equations.
of kidney function.
• Fall in GFR leads to accumulation of waste products
of metabolism in blood. GFR <15 ml/min per 1.73 m2 Clearance Tests to Measure Glomerular
is associated with uremia. Filtration Rate (GFR)
Glomerular filtration rate refers to the rate in ml/min at
which a substance is cleared from the circulation by the
Normal GFR in young adults is 120-130 ml/min per 1.73 glomeruli. The ability of the glomeruli to filter a substance
m2 of body surface area. GFR declines progressively with from the blood is assessed by clearance studies. If a
age (due to arteriolosclerosis of glomeruli). After 40 years substance is not bound to protein in plasma, is completely
of age, there is a steady and progressive fall in the GFR filtered by the glomeruli, and is neither secreted nor
at the rate of 1 ml/minute/year because of reduction in reabsorbed by the tubules, then its clearance rate is equal
the number of glomeruli due to arteriolosclerosis. to the glomerular filtration rate (GFR). Clearance of a
GFR is measured to (i) detect suspected incipient substance refers to the volume of plasma, which is
kidney disease (i.e. early detection), (ii) monitor course completely cleared of that substance per minute; it is
of established kidney disease, (iii) plan renal replacement calculated from the following formula:
therapy in advanced renal disease, and (iv) adjust dosage
of certain drugs which are nephrotoxic. UV
Clearance = ——
Based on GFR, chronic kidney disease is divided into P
following stages (US National Kidney Foundation where, U = concentration of a substance in urine in
Kidney Disease Quality Outcomes Initiative Classifi- mg/dl; V = volume of urine excreted in ml/min; and P
cation of Chronic Kidney Disease, 2002): = concentration of the substance in plasma in mg/dl.
• Stage 1: Kidney damage with normal or increased Since U and P are in the same units, they cancel each
GFR (GFR ≥ 90 ml/min/1.73 m2) other and the clearance value is expressed in the same
• Stage 2: Kidney damage with mildly reduced GFR unit as V i.e. ml/min. All clearance values are adjusted
(GFR 60-89 ml/min/1.73 m2) to a standard body surface area i.e. 1.73 m2.

tahir99 - UnitedVRG
vip.persianss.ir
32 Essentials of Clinical Pathology

The agents used for measurement of GFR are: and is not returned to circulation after filtration. It is a
• Exogenous: Inulin, Radiolabelled ethylenediamine more sensitive and specific marker of impaired renal
tetraacetic acid (51Cr- EDTA), 125I-iothalamate function than plasma creatinine. Its level is not affected
• Endogenous: Creatinine, Urea, Cystatin C by sex, diet, or muscle mass. It is thought that cystatin C
The agent used for measurement of GFR should have is a superior marker for estimation of GFR than creatinine
following properties: (1) It should be physiologically inert clearance. It is measured by immunoassay.
and preferably endogenous, (2) It should be freely filtered
by glomeruli and should be neither reabsorbed nor Creatinine Clearance
secreted by renal tubules, (3) It should not bind to plasma
This is the most commonly used test for measuring GFR.
proteins and should not be metabolized by kidneys, and
Creatinine is being produced constantly from creatine
(4) It should be excreted only by the kidneys. However,
in muscle. It is completely filtered by glomeruli and is
there is no such ideal endogenous agent.
not reabsorbed by tubules; however, a small amount is
Clearance tests are cumbersome to perform,
secreted by tubules.
expensive, and not readily available. One major
problem with clearance studies is incomplete urine A 24-hour urine sample is preferred to overcome the
collection. problem of diurnal variation of creatinine excretion and
Abnormal clearance occurs in: (i) pre-renal factors: to reduce the inaccuracy in urine collection.
reduced blood flow due to shock, dehydration, and After getting up in the morning, the first voided urine
congestive cardiac failure; (ii) renal diseases; and is discarded. Subsequently all the urine passed is
(iii) obstruction to urinary outflow. collected in the container provided. After getting up in
the next morning, the first voided urine is also collected
Inulin Clearance and the container is sent to the laboratory. A blood
sample for estimation of plasma creatinine is obtained
Inulin, an inert plant polysaccharide (a fructose polymer), at midpoint of urine collection. Creatinine clearance is
is filtered by the glomeruli and is neither reabsorbed nor calculated from (1) concentration of creatinine in urine
secreted by the tubules; therefore it is an ideal agent for in mg/ml (U), (2) volume of urine excreted in ml/min
measuring GFR. A bolus dose of inulin (25 ml of 10% (V) (this is calculated by the formula: volume of urine
solution IV) is administered followed by constant collected/collection time in minutes e.g. volume of urine
intravenous infusion (500 ml of 1.5% solution at the rate collected in 24 hours ÷ 1440), and (3) concentration of
of 4 ml/min). Timed urine samples are collected and creatinine in plasma in mg/dl (P). Creatinine clearance
blood samples are obtained at the midpoint of timed in ml/min per 1.73 m2 is then derived from the formula
urine collection. This test is considered as the ‘gold UV/P.
standard’ (or reference method) for estimation of GFR. Because of secretion of creatinine by renal tubules,
However, this test is rarely used because it is time the above formula overestimates GFR by about 10%. In
consuming, expensive, constant intravenous infusion of advanced renal failure, secretion of creatinine by tubules
inulin is needed to maintain steady plasma level, and is increased and thus overestimation of GFR is even more.
difficulties in laboratory analysis. Average inulin Jaffe’s reaction (see later under serum creatinine) used
clearance for males is 125 ml/min/1.73 m2 and for for estimation of creatinine measures creatinine as well
females is 110 ml/min/1.73 m2. In children less than 2 as some other substances (non-creatinine chromogens)
years and in older adults, clearance is low. This test is in blood and thus gives slightly higher result. Thus effect
largely limited to clinical research. of tubular secretion of creatinine is somewhat balanced
by slight overestimation of serum creatinine by Jaffe’s
Clearance of Radiolabeled Agents reaction.
Urinary clearance of radiolabeled iothalamate (125I- To provide values closer to the actual GFR, cimetidine
iothalamate) correlates closely with inulin clearance. (which blocks secretion by renal tubules) can be
However, this method is expensive with risk of exposure administered before commencing urine collection
to radioactive substances. Other radiolabelled substances (cimetidine-enhanced creatinine clearance).
used are 51Cr-EDTA and 99Tc-DTPA. Creatinine clearance is not an ideal test for estimation
of GFR because of following reasons:
Cystatin C Clearance 1. A small amount of creatinine is secreted by renal
This is a cysteine protease inhibitor of MW 13,000, which tubules that increase even further in advanced renal
is produced at a constant rate by all the nucleated cells. failure.
It is not bound to protein, is freely filtered by glomeruli 2. Collection of urine is often incomplete.

tahir99 - UnitedVRG
vip.persianss.ir
Renal Function Tests 33

3. Creatinine level is affected by intake of meat and It is recommended by National Kidney Foundation
muscle mass. (USA) to calculate creatinine clearance by Cockcroft and
4. Creatinine level is affected by certain drugs like Gault or other equation from serum creatinine value
cimetidine, probenecid, and trimethoprim (which rather than estimating creatinine clearance from a 24-
block tubular secretion of creatinine). hour urine sample. This is because the latter test is
inconvenient, time-consuming, and often inaccurate.
Urea Clearance
Blood Biochemistry
Urea is filtered by the glomeruli, but about 40% of the
filtered amount is reabsorbed by the tubules. The Two biochemical parameters are commonly used to
reabsorption depends on the rate of urine flow. Thus it assess renal function: blood urea nitrogen (BUN) and
underestimates GFR, depends on the urine flow rate, and serum creatinine. Although convenient, they are
is not a sensitive indicator of GFR. insensitive markers of glomerular function.
BUN and serum creatinine, by themselves, are not
Blood Urea Nitrogen (BUN)
sensitive indicators of early renal impairment since
values may be normal e.g. if baseline values of serum Urea is produced in the liver from amino acids (ingested
creatinine is 0.5 mg/dl, then 50% reduction in kidney or tissue-derived). Amino acids are utilized to produce
function would increase it to 1.0 mg/dl. Thus clearance energy, synthesize proteins, and are catabolized to
tests are more helpful in early cases. If biochemical tests ammonia. Urea is produced in the liver from ammonia
are normal and renal function impairment is suspected, in the Krebs urea cycle. Ammonia is toxic and hence is
then creatinine clearance test should be carried out. If converted to urea, which is then excreted in urine
biochemical tests are abnormal, then clearance tests need (Fig. 2.1).
not be done. The concentration of blood urea is usually expressed
as blood urea nitrogen. This is because older methods
Estimation of Creatinine Clearance from Serum estimated only the nitrogen in urea. Molecular weight
Creatinine by Prediction Equations of urea is 60, and 28 grams of nitrogen are present in a
gm mole of urea. As the relationship between urea and
One can estimate GFR from age, sex, body weight, and BUN is 60/28, BUN can be converted to urea by
serum creatinine value of a person from the following multiplying BUN by 2.14, i.e. the real concentration of
formula (Cockcroft and Gault): urea is BUN × (60/28).
Creatinine clearance
Urea is completely filtered by the glomeruli, and
(140 - Age in years) × (Body weight in kg) about 30-40% of the filtered amount is reabsorbed in the
in ml/min =
(72 × Serum creatinine in mg/dl) renal tubules depending on the person’s state of
hydration
In females, the value obtained from above equation Blood level of urea is affected by a number of non-
is multiplied by 0.85 to get the result. renal factors (e.g. high protein diet, upper gastrointestinal

Fig. 2.1: Formation of urea from protein breakdown


34 Essentials of Clinical Pathology

hemorrhage, liver function, etc.) and therefore utility of • It is not reabsorbed, and very little is secreted by
BUN as an indicator of renal function is limited. Also tubules.
considerable destruction of renal parenchyma is required With muscle mass remaining constant, increased
before elevation of blood urea can occur. creatinine level reflects reduction of glomerular filtration
The term azotemia refers to the increase in the blood rate. However, because of significant kidney reserve,
level of urea; uremia is the clinical syndrome resulting increase of serum creatinine level (from 1.0 mg/dl to 2.0
from this increase. If renal function is absent, BUN rises mg/dl) in blood does not occur until about 50% of kidney
by 10-20 mg/dl/day. function is lost. Therefore, serum creatinine is not a
sensitive indicator of early renal impairment. Also,
Causes of increased BUN: laboratory report showing serum creatinine “within
1. Pre-renal azotemia: shock, congestive heart failure, normal range” does not necessarily mean that the level
salt and water depletion is normal; the level should be correlated with body
2. Renal azotemia: impairment of renal function weight, age, and sex of the individual. If renal function
3. Post-renal azotemia: obstruction of urinary tract is absent, serum creatinine rises by 1.0 to 1.5 mg/dl/day
4. Increased rate of production of urea: (Fig. 2.2).
• High protein diet
• Increased protein catabolism (trauma, burns,
fever)
• Absorption of amino acids and peptides from a
large gastrointestinal hemorrhage or tissue
hematoma

Methods for estimation of BUN:


Two methods are commonly used.
1. Diacetyl monoxime urea method: This is a direct
method. Urea reacts with diacetyl monoxime at high
temperature in the presence of a strong acid and an
oxidizing agent. Reaction of urea and diacetyl
monoxime produces a yellow diazine derivative. The
intensity of color is measured in a colorimeter or
spectrophotometer.
2. Urease- Berthelot reaction: This is an indirect method.
Enzyme urease splits off ammonia from the urea
molecule at 37°C. Ammonia generated is then reacted Fig. 2.2: Relationship between glomerular filtration rate and
serum creatinine. Significant increase of serum creatinine does
with alkaline hypochlorite and phenol with a catalyst
not occur till a considerable fall in GFR
to produce a stable color (indophenol). Intensity of
color produced is then measured in a spectro-
Causes of Increased Serum Creatinine Level
photometer at 570 nm.
Reference range for BUN in adults is 7-18 mg/dl. In 1. Pre-renal, renal, and post-renal azotemia
adults > 60 years, level is 8-21 mg/dl. 2. Large amount of dietary meat
3. Active acromegaly and gigantism
Serum Creatinine Causes of Decreased Serum Creatinine Level
Creatinine is a nitrogenous waste product formed in 1. Pregnancy
muscle from creatine phosphate. Endogenous production 2. Increasing age (reduction in muscle mass)
of creatinine is proportional to muscle mass and body
weight. Exogenous creatinine (from ingestion of meat) Methods for Estimation of Serum Creatinine
has little effect on daily creatinine excretion. The test for serum creatinine is cheap, readily available,
Serum creatinine is a more specific and more sensitive and simple to perform. There are two methods that are
indicator of renal function as compared to BUN because: commonly used:
• It is produced from muscles at a constant rate and its 1. Jaffe’s reaction (Alkaline picrate reaction):
level in blood is not affected by diet, protein This is the most widely used method. Creatinine
catabolism, or other exogenous factors; reacts with picrate in an alkaline solution to produce
Renal Function Tests 35

a yellow-red color. The color is measured in a Causes of Decreased BUN/Creatinine Ratio (<10:1)
spectrophotometer at 485 nm. Certain substances in
• Acute tubular necrosis
plasma (such as glucose, protein, fructose, ascorbic
• Low protein diet, starvation
acid, acetoacetate, acetone, and cephalosporins) react • Severe liver disease
with picrate in a similar manner; these are called as
non-creatinine chromogens (and can cause false Microalbuminuria and Albuminuria
elevation of serum creatinine level). Thus ‘true’
Normally, a very small amount of albumin is excreted
creatinine is less by 0.2 to 0.4 mg/dl when estimated
in urine. The earliest evidence of glomerular damage in
by Jaffe’s reaction. diabetes mellitus is occurrence of microalbuminuria
2. Enzymatic methods: These methods use enzymes (albuminuria in the range of 30 to 300 mg/24 hours). An
that cleave creatinine; hydrogen peroxide produced albuminuria > 300-mg/24 hour is termed clinical or overt
then reacts with phenol and a dye to produce a and indicates significant glomerular damage. (See
colored product, which is measured in a spectro- “Proteinuria” under Chapter 1 “Examination of Urine”).
photometer.
Tests to Evaluate Tubular Function
Reference range:
Adult males: 0.7-1.3 mg/dl. Tests to Assess Proximal Tubular Function
Adult females: 0.6-1.1 mg/dl. Renal tubules efficiently reabsorb 99% of the glomerular
Serum creatinine alone should not be used to assess filtrate to conserve the essential substances like glucose,
renal function. This is because serum creatinine amino acids, and water.
concentration depends on age, sex, muscle mass, 1. Glycosuria: In renal glycosuria, glucose is excreted in
glomerular filtration and amount of tubular secretion. urine, while blood glucose level is normal. This is
Thus, normal serum creatinine range is wide. Serum because of a specific tubular lesion which leads to
creatinine begins to rise when GFR falls below 50% of impairment of glucose reabsorption. Renal glycosuria
normal. Minor rise of serum creatinine is associated with is a benign condition. Glycosuria can also occur in
significant reduction of GFR (Fig 2.2). Therefore early Fanconi syndrome.
stage of chronic renal impairment cannot be detected by 2. Generalized aminoaciduria: In proximal renal tubular
measurement of serum creatinine alone. dysfunction, many amino acids are excreted in urine
due to defective tubular reabsorption.
BUN/Serum Creatinine Ratio 3. Tubular proteinuria (Low molecular weight proteinuria):
Clinicians commonly calculate BUN/creatinine ratio to Normally, low molecular weight proteins (β 2 –
discriminate pre-renal and post-renal azotemia from microglobulin, retinol-binding protein, lysozyme, and
α1-microglobulin) are freely filtered by glomeruli and
renal azotemia. Normal ratio is 12:1 to 20:1.
are completely reabsorbed by proximal renal tubules.
With tubular damage, these low molecular weight
Causes of Increased BUN/Creatinine Ratio (>20:1): proteins are excreted in urine and can be detected by
1. Increased BUN with normal serum creatinine: urine protein electrophoresis. Increased amounts of
• Pre-renal azotemia (reduced renal perfusion) these proteins in urine are indicative of renal tubular
damage.
• High protein diet
• Increased protein catabolism 4. Urinary concentration of sodium: If both BUN and serum
creatinine are acutely increased, it is necessary to
• Gastrointestinal hemorrhage
distinguish between prerenal azotemia (renal
2. Increase of both BUN and serum creatinine with underperfusion) and acute tubular necrosis. In
disproportionately greater increase of BUN: prerenal azotemia, renal tubules are functioning
• Post-renal azotemia (Obstruction to the outflow normally and reabsorb sodium, while in acute tubular
of urine) necrosis, tubular function is impaired and sodium
Obstruction to the urine outflow causes diffusion absorption is decreased. Therefore, in prerenal
of urinary urea back into the blood from tubules azotemia, urinay sodium concentration is < 20 mEq/
because of backpressure. L while in acute tubular necrosis, it is > 20 mEq/L.
36 Essentials of Clinical Pathology

5. Fractional excretion of sodium (FENa): Measurement of 2. Urine osmolality: The most commonly employed test
urinary sodium concentration is affected by urine to evaluate tubular function is measurement of urine/
volume and can produce misleading results. There- plasma osmolality. This is the most sensitive method for
fore, to avoid this, fractional excretion of sodium is determination of ability of concentration. Osmolality
calculated. This refers to the percentage of filtered measures number of dissolved particles in a solution.
sodium that has been absorbed and percentage that Specific gravity, on the other hand, is the ratio of mass of
has been excreted. Measurement of fractional sodium a solution to the mass of water i.e. it measures total mass
excretion is a better indicator of tubular absorption of solute. Specific gravity depends on both the number
of sodium than quantitation of urine sodium alone. and the nature of dissolved particles while osmolality is
This test is indicated in acute renal failure. In oliguric exact number of solute particles in a solution. Specific
patients, this is the most reliable means of early gravity measurement can be affected by the presence of
distinction between pre-renal failure and renal failure solutes of large molecular weight like proteins and
due to acute tubular necrosis. It is calculated from the glucose, while osmolality is not. Therefore measurement
following formula: of osmolality is preferred.
When solutes are dissolved in a solvent, certain
(Urine sodium × Plasma creatinine)
× 100% changes take place like lowering of freezing point,
(Plasma sodium × Urine creatinine) increase in boiling point, decrease in vapor pressure, or
increase of osmotic pressure of the solvent. These
In pre-renal failure this ratio is less than 1%, and in
properties are made use of in measuring osmolality by
acute tubular necrosis it is more than 1%. In pre-renal
an instrument called as osmometer.
failure (due to reduced renal perfusion), aldosterone
Osmolality is expressed as milliOsmol/kg of water.
secretion is stimulated which causes maximal sodium
Urine/plasma osmolality ratio is helpful in distin-
conservation by the tubules and the ratio is less than 1%.
guishing pre-renal azotemia (in which ratio is higher)
In acute tubular necrosis, maximum sodium reabsorption
from acute renal failure due to acute tubular necrosis (in
is not possible due to tubular cell injury and consequently
which ratio is lower). If urine and plasma osmolality are
the ratio will be more than 1%. Values above 3% are almost similar, then there is defective tubular reabsorp-
strongly suggestive of acute tubular necrosis. tion of water.
Tests to Assess Distal Tubular Function 3. Water deprivation test If the value of baseline osmolality
of urine is inconclusive, then water deprivation test is
1. Urine specific gravity: Normal specific gravity is 1.003 performed. In this test, water intake is restricted for a
to 1.030. It depends on state of hydration and fluid intake. specified period of time followed by measurement of
i. Causes of increased specific gravity: specific gravity or osmolality. Normally, urine osmolality
a. Reduced renal perfusion (with preservation of should rise in response to water deprivation. If it fails to
concentrating ability of tubules), rise, then desmopressin is administered to differentiate
b. Proteinuria, between central diabetes insipidus and nephrogenic
c. Glycosuria, diabetes insipidus. Urinary concentration ability is
d. Glomerulonephritis. corrected after administration of desmopressin in central
e. Urinary tract obstruction. diabetes insipidus, but not in nephrogenic diabetes
ii. Causes of reduced specific gravity: insipidus.
a. Diabetes insipidus If urine osmolality is > 800 mOsm/kg of water or
b. Chronic renal failure specific gravity is ≥1.025 following dehydration,
c. Impaired concentrating ability due to diseases of concentrating ability of renal tubules is normal. However,
tubules. normal result does not rule out presence of renal disease.
As a test of renal function, it gives information about False result will be obtained if the patient is on low-salt,
the ability of renal tubules to concentrate the glomerular low-protein diet or is suffering from major electrolyte
and water disturbance.
filtrate. This concentrating ability is lost in diseases of
renal tubules. 4. Water loading antidiuretic hormone suppression test This
Fixed specific gravity of 1.010, which cannot be test assesses the capacity of the kidney to make urine
lowered or increased by increasing or decreasing the fluid dilute after water loading.
intake respectively, is an indication of chronic renal After overnight fast, patient empties the bladder and
failure. drinks 20 ml/kg of water in 15-30 minutes. The urine is
Renal Function Tests 37

collected at hourly intervals for the next 4 hours for Indications for Renal Biopsy
measurements of urine volume, specific gravity, and
1. Nephrotic syndrome in adults (most common
osmolality. Plasma levels of antidiuretic hormone and
indication)
serum osmolality should be measured at hourly intervals.
2. Nephrotic syndrome not responding to cortico-
Normally, more than 90% of water should be excreted
steroids in children.
in 4 hours. The specific gravity should fall to 1.003 and
3. Acute nephritic syndrome for differential diagnosis
osmolality should fall to < 100 mOsm/kg. Plasma level
4. Unexplained renal insufficiency with near-normal
of antidiuretic hormone should be appropriate for serum
kidney dimensions on ultrasonography
osmolality. In renal function impairment, urine volume
5. Asymptomatic hematuria, when other diagnostic
is reduced (<80% of fluid intake is excreted) and specific
tests fail to identify the source of bleeding
gravity and osmolality fail to decrease. The test is also
6. Isolated non-nephrotic range proteinuria (1-3 gm/
impaired in adrenocortical insufficiency, malabsorption,
24 hours) with renal impairment
obesity, ascites, congestive heart failure, cirrhosis, and
7. Impaired function of renal graft
dehydration.
8. Involvement of kidney in systemic disease like
This test is not advisable in patients with cardiac
systemic lupus erythematosus or amyloidosis
failure or kidney disease. If there is failure to excrete
water load, fatal hyponatremia can occur.
Contraindications
5. Ammonium chloride loading test (Acid load test): Diagnosis
1. Uncontrolled severe hypertension
of renal tubular acidosis is usually considered after
2. Hemorrhagic diathesis
excluding other causes of metabolic acidosis. This test is
3. Solitary kidney
considered as a ‘gold standard’ for the diagnosis of distal
4. Renal neoplasm (to avoid spread of malignant cells
or type 1 renal tubular acidosis. Urine pH and plasma
along the needle track)
bicarbonate are measured after overnight fasting. If pH
5. Large and multiple renal cysts
is less than 5.4, acidifying ability of renal tubules is
6. Small, shrunken kidneys
normal. If pH is greater than 5.4 and plasma bicarbonate
7. Acute urinary tract infection like pyelonephritis
is low, diagnosis of renal tubular acidosis is confirmed.
8. Urinary tract obstruction
In both the above cases, further testing need not be
performed. In all other cases in which neither of above
Complications
results is obtained, further testing is carried out. Patient
is given ammonium chloride orally (0.1 gm/kg) over 1 1. Hemorrhage: As renal cortex is highly vascular, major
hour after overnight fast and urine samples are collected risk is bleeding in the form of hematuria or peri-
hourly for next 6-8 hours. Ammonium ion dissociates nephric hematoma. Severe bleeding may occasionally
into H+ and NH3. Ammonium chloride makes blood necessitate blood transfusion and rarely removal of
acidic. If pH is less than 5.4 in any one of the samples, kidney.
acidifying ability of the distal tubules is normal. 2. Arteriovenous fistula
3. Infection
RENAL BIOPSY 4. Accidental biopsy of another organ or perforation of
viscus (liver, spleen, pancreas, adrenals, intestine, or
Renal biopsy refers to obtaining a small piece of kidney gallbladder)
tissue for microscopic examination. Percutaneous renal 5. Death (rare).
biopsy was first performed by Alwall in 1944. In renal
disease, renal biopsy is helpful to: Procedure
• Establish the diagnosis 1. Patient’s informed consent is obtained.
• Assess severity and activity of disease 2. Ultrasound/CT scan is done to document the
• Assess prognosis by noting the amount of scarring location and size of kidneys.
• To plan treatment and monitor response to therapy 3. Blood pressure should be less than 160/90 mm of
Renal biopsy is associated with the risk of procedure- Hg. Bleeding time, platelet count, prothrombin
related morbidity and rarely mortality. Therefore, before time, and activated partial thromboplastin time
performing renal biopsy, risks of the procedure and should be normal. Blood sample should be drawn
benefits of histologic examination should be evaluated for blood grouping and cross matching, as blood
in each patient. transfusion may be needed.
38 Essentials of Clinical Pathology

4. Patient is sedated before the procedure. REFERENCE RANGES


5. Patient lies in prone position and kidney is
identified with ultrasound. Inulin clearance:
6. The skin over the selected site is disinfected and a Men: 90-139 ml/min/1.73 m2
local anesthetic is infiltrated. Women: 80-125 ml/min/1.73 m2
7. A small skin incision is given with a scalpel (to Creatinine clearance:
insert the biopsy needle). Localization of kidney Men: 110-150 ml/min/1.73 m2
is done with a fine bore 21 G lumbar puncture Women: 105-132 ml/min/1.73 m2
needle. A local anesthetic is infiltrated down to Blood urea nitrogen (BUN):
the renal capsule. Adults: 7-18 mg/dl
8. A tru-cut biopsy needle or spring loaded biopsy >60 years: 8-21 mg/dl
gun is inserted under ultrasound guidance and
advanced down to the lower pole. Biopsy is Serum creatinine:
usually obtained from lateral border of lower pole. Adult males: 0.7-1.3 mg/dl
Patient should hold his/her breath in full Adult females: 0.6-1.1 mg/dl
inspiration during biopsy. After obtaining the Children (<20 years):
biopsy and removal of needle, patient is allowed Boys: 0.35 + Age in years/40
to breath normally. Girls: 0.35 + Age in years/55
9. The biopsy should be placed in a drop of saline BUN/Serum creatinine ratio: 12: 1 to 20:1
and examined under a dissecting microscope for
adequacy.
CRITICAL VALUES
10. Patient is turned to supine position. Vital signs
and appearance of urine should be monitored at Serum creatinine: > 5 mg/dl
regular intervals. Patient is usually kept in the BUN: > 80 mg/dl
hospital for 24 hours.
Kidney biopsy can be divided into three parts for light
microscopy, immunofluorescence, and electron micro- BIBLIOGRAPHY
scopy. For light microscopy, renal biopsy is routinely 1. Gaw A, Murphy MJ, Cowan RA, O’Reilly DSJ, Stewart
fixed in neutral buffered formaldehyde. Sections are MJ, Shepherd J. Clinical Biochemistry: An Illustrated
stained by: Colour Text (3rd Ed). Edinburgh: Churchill Livingstone
• Hematoxylin and eosin (for general architecture of 2004.
kidney and cellularity) 2. Johnson CA, Levey AS, Coresh J, Levin A, Lau J, Eknoyan
• Periodic acid Schiff: To highlight basement mem- G. Clinical practice guidelines for chronic kidney disease
brane and connective tissue matrix. in adults: Part II. Glomerular filtration rate, proteinuria,
• Congo red: For amyloid. and other markers Am Fam Physician 2004;70:1091-7.
For electron microscopy, tissue is fixed in glutaraldeyde. 3. Stevens LA, Coresh J, Green T, Levey AS. Assessing
In immunohistochemistry, tissue deposits of IgG, IgA, kidney function-measured and estimated glomerular
IgM, C3, fibrin, and κ and λ light chains can be detected filtration rate. N Engl J Med 2006;354:2473-83.
by using appropriate antibodies. Many kidney diseases 4. Stevens LA, Levey AS. Measurement of kidney function.
are immune-complex mediated. Med Clin N Am 2005;89:457-73.
3

Diabetes Mellitus

Diabetes mellitus (DM) is a metabolic group of disorders


characterized by persistent hyperglycemia due to
deficiency and/or diminished effectiveness of insulin.
There are derangements of carbohydrate, protein, and
fat metabolism due to failure of insulin action on target
cells. Typical features of DM are as follows:
• Fasting hyperglycemia
• Glycosuria
• Symptoms due to marked hyperglycemia: polyuria,
polydipsia, weight loss, weakness, polyphagia, and
blurred vision
• Long-term complications like atherosclerosis (leading
to ischemic heart disease, cerebrovascular disease,
and peripheral vascular disease) and microangio-
pathy (which can cause nephropathy with risk of
renal failure; retinopathy with potential loss of vision;
and peripheral neuropathy with risk of foot ulcers,
amputations, or Charcot joints).
• Acute metabolic complications (hyperosmolar
hyperglycemic state, diabetic ketoacidosis).
• Susceptibility to infections especially of skin,
respiratory tract, and urinary tract.

METABOLIC ACTIONS OF INSULIN Fig. 3.1: Proinsulin, insulin, and C-peptide. The biochemical
cleavage of proinsulin to insulin and C-peptide occurs in Golgi
Insulin is the major hormone regulating blood glucose apparatus of β cell. Secretion of insulin is stimulated by glucose,
level. Insulin is synthesized by β cells of pancreas as mannose, amino acids, and sulfonylureas
preproinsulin, which is rapidly converted to proinsulin.
Proinsulin is a single chain polypeptide. In the Golgi
apparatus, proinsulin is broken down into 2 units- insulin “Stress hormones” like glucagons, glucocorticoids,
(51 amino acids) and C (connecting)-polypeptide (31 growth hormone, and adrenaline oppose action of
amino acids) (Fig. 3.1). Both insulin and C peptide are insulin.
stored in membrane-bound granules in the cytoplasm
of β cells. Upon stimulation (mainly by blood glucose), CLASSIFICATION OF DIABETES MELLITUS
both insulin and C peptide are released in circulation. According to American Diabetes Association (1997), DM
C peptide is often measured as a marker of activity of β is classified into following types:
cells. C peptide has no known function. • Type 1 (Absolute deficiency of insulin due to
Insulin acts on various cells (especially those of liver, destruction of β cells of pancreas)
muscle, and adipose tissue) through receptors. – Immune mediated
Important actions of insulin are shown in Box 3.1. – Idiopathic
40 Essentials of Clinical Pathology

Box 3.1: Major actions of insulin


• Increases:
– Uptake of glucose in skeletal muscle and adipose
tissue
– Glycogenesis in liver and muscle
– Fatty acid and triglycerides in liver and adipose
tissue
– Protein synthesis in liver and muscle
• Decreases:
– Gluconeogenesis in liver
– Glycogenolysis in liver and muscle
– Lipolysis in adipose tissue
– Ketogenesis in liver

• Type 2 (Insulin resistance along with relative


deficiency of insulin secretion)
• Other specific types
• Gestational DM (onset or first recognition of glucose
intolerance during pregnancy).
Fig. 3.2: Pathogenesis of type I diabetes mellitus
Type 1 Diabetes Mellitus
It accounts for 5-10% of all cases of DM. This was
infiltration by cytotoxic CD8+ T lymphocytes in and
previously called as insulin-dependent DM or IDDM
around islets. It is thought that many cases follow a viral
(because insulin therapy is essential to prevent ketosis),
juvenile-onset DM (because it commonly presents during infection that has damaged the islet cells of pancreas (Fig.
childhood or adolescence), brittle DM, or ketosis-prone 3.2). Markers of immune destruction of β cells, which
DM. It is characterized by absolute deficiency of insulin can be detected in peripheral blood, are islet cell
secretion. antibodies, autoantibodies to insulin, autoantibodies to
Cell-mediated autoimmune destruction of β cells glutamic acid decarboxylase (GAD65), and autoanti-
of pancreas is responsible for majority of cases of type bodies to tyrosine phosphatases (IA-2 and IA-2b). The
1 DM (immune-mediated type 1 DM), leading to disease has strong association with HLA DR3 and HLA
inability of pancreas to synthesize insulin. There is DR4 haplotypes (Fig. 3.3). This type occurs mainly in

Fig. 3.3: HLA-linked genetic predisposition to type 1 diabetes mellitus


Diabetes Mellitus 41

children and adolescents, but can occur at any age. These Other Specific Types
patients are also at risk of other autoimmune disorders
There are several forms of DM associated with under-
like Graves’ disease, Hashimoto’s thyroiditis, vitiligo,
lying conditions:
Addison’s disease, pernicious anemia, etc.
• Genetic defects of β cell function: In these disorders,
Some cases of type 1 DM do not have any known
insulin secretion from β cells is impaired. These are
etiologies or evidence of autoimmunity. These indivi-
duals are of Asian or African origin and their disease is called as maturity onset diabetes of the young
strongly inherited. This form of type 1 DM is called as (MODY). They are inherited in an autosomal
idiopathic DM. dominant manner and they are caused by mutations
in genetic loci such as hepatic nuclear factor,
Type 2 Diabetes Mellitus glucokinase, etc.
• Genetic defects in insulin action: These result from
This is the most common form of DM comprising about mutations in insulin receptor gene.
90-95% of all patients of DM. This was previously called • Diseases of exocrine pancreas: Diseases causing
as non-insulin-dependent DM (NIDDM), maturity-onset generalized pancreatic damage can result in DM.
DM (because onset usually occurs during adult life), These include cystic fibrosis, hemochromatosis,
stable DM, or ketosis-resistant DM. It is characterized chronic pancreatitis, trauma, pancreatectomy, and
by insulin resistance along with relative deficiency of pancreatic cancer.
insulin secretion (i.e. inadequate insulin secretory • Endocrine disorders: Several hormones inhibit the
action of insulin. Excessive secretion of these
response to overcome peripheral insulin resistance).
hormones will cause DM. Hyperglycemia is corrected
(Fig. 3.4). Type 2 DM is not HLA-linked and there is no
following resolution of the primary endocrinopathy.
role of autoimmunity in its pathogenesis. It has a strong Endocrine disorders associated with hyperglycemia
genetic predisposition. Type 2 DM occurs more are:
frequently in individuals with positive family history – Acromegaly: Excess growth hormone.
(parents or siblings with DM), obesity (≥ 20% over ideal – Cushing’s syndrome: Excess cortisol.
body weight or body mass index ≥ 25 kg/m2), hyper- – Glucagonoma: Excess glucagon
tension (>140/90 mm Hg in adults), dyslipidemia, lack – Pheochromocytoma: Excess epinephrine.
of physical activity, pre-diabetes (impaired fasting – Hyperthyroidism: Excess thyroxine
glucose or impaired glucose tolerance), and prior • Drug- or chemical-induced DM: Drugs or chemicals can
gestational DM. impair insulin secretion or insulin action. Destruction
Type 2 diabetes is more common in certain racial of β cells and formation of islet cell antibodies have
also been reported with some drugs. Examples
groups like South Asians and Africans. Rising trend of
include thiazide diuretics, α-interferon, and gluco-
type 2 DM is due to increasing tendency towards obesity
corticoids.
in urban populations coupled with high-calorie diet.
• Infections: Certain viral infections (such as Coxsackie
Differences between type 1 and type 2 DM are listed virus B, congenital rubella, cytomegalovirus) can
in Table 3.1. cause destruction of β cells.
• Other genetic syndromes sometimes associated with DM:
Many genetic syndromes (e.g. Down’s syndrome,
Klinefelter’s syndrome, Turner’s syndrome) are
associated with increased risk of developing DM.

Gestational Diabetes Mellitus (GDM)


This refers to the onset or first recognition of glucose
intolerance during pregnancy. GDM occurs in 2-3% of
all pregnant women. It is associated with increased risk
of high birth weight of the newborn, cardiac defects,
polyhydramnios, intrauterine fetal loss (due to
placental insufficiency), premature birth, hypertension
during pregnancy, pre-eclampsia, and alteration in
Fig. 3.4: Pathogenesis of type 2 diabetes mellitus duration of pregnancy. Early diagnosis and treatment
42 Essentials of Clinical Pathology

Table 3.1: Differences between type 1 and type 2 diabetes mellitus


Parameter Type 1 DM Type 2 DM

1. Previous names Type I, IDDM, Juvenile-onset, Type II, NIDDM, maturity-onset,


Ketosis-prone, Brittle Ketosis-resistant, Stable
2. % of all DM cases 5-10% 90-95%
3. Age of onset < 35 years > 35 years
4. Obesity No (weight loss common) Common
5. Presentation Acute; symptoms (classical) Insidious; symptoms since few
since few weeks months or years
6. Genetic predisposition Low Strong
7. Glucose intolerance Marked Mild
8. Ketoacidosis Common Uncommon
9. Hyperosmolar hyperglycemic state Uncommon Common
10. Serum insulin Undetectable Normal or high
11. Autoimmunity (Islet cell antibodies) Majority of cases No
12. HLA-linkage Yes (DR3, DR4) No
13. Pathogenesis Absolute insulin deficiency Insulin resistance with relative
insulin deficiency
14. Serum C-peptide level Very low to absent Normal or high
15. Predisposing factors Viral infections, toxins Obesity, lack of physical activity
16. Insulin requirement for treatment Always In some situations

of GDM are essential to prevent perinatal morbidity and acute complications of DM and microangiopathy, but are
mortality. After delivery, GDM can have following at increased risk of cardiovascular disease (1.5 times
course: normal individuals). Development of DM can be delayed
1. Return to normal glucose tolerance (however, many or prevented through modest amount of weight loss, diet
of these patients are likely to develop DM during modification, and regular moderate exercise in patients
subsequent years), or with prediabetes.
2. Persistence of DM or impaired glucose tolerance.
Patient should be reassessed 6 weeks or later METABOLIC SYNDROME (INSULIN
following delivery. RESISTANCE SYNDROME, REAVEN’S
Risk factors for GDM are shown in Box 3.2. SYNDROME, SYNDROME X)
PREDIABETES The metabolic syndrome refers to a constellation of lipid
Prediabetes is a state in which plasma glucose level is and non-lipid risk factors that are of metabolic origin and
higher than normal but not high enough for diagnosis of associated with risk of cardiovascular disease. The Adult
DM. It is also referred to as impaired fasting glucose (IFG) Treatment Panel III (ATP III) 1 of the National Cholesterol
or impaired glucose tolerance (IGT), depending on which Education Program in 2001 proposed criteria for
test is used for its detection. Studies have shown that diagnosing the metabolic syndrome. The metabolic
majority of individuals with prediabetes develop type 2 syndrome is diagnosed when three or more out of
DM within 10 years. Prediabetic persons do not develop following five criteria are present.
• Abdominal obesity
– Men: Waist circumference > 40 inches (102 cm)
– Women: Waist circumference > 35 inches (88 cm)
Box 3.2: Risk factors for gestational DM • Fasting glucose ≥ 110 to < 126 mg/dl (As these criteria
• Past history of GDM were proposed in 2001, the fasting plasma glucose
• Previous high-birth-weight baby
value should be reduced to 100 mg/dl according to
revised criteria proposed by American Diabetes
• Obesity
Association in 2004).
• Family history of diabetes mellitus
• Blood pressure ≥ 130/> 85 mm Hg
• High-risk ethnic group: South Asian or African.
• Triglycerides ≥ 150 mg/dl (> 1.7 mmol/L)
Diabetes Mellitus 43

• Plasma high density lipoprotein (HDL)-cholesterol insulin deficiency). In reaction to this, there are
– Men: < 40 mg/dl (< 1 mmol/L) compensatory glycogenolysis (breakdown of glyco-
– Women: < 50 mg/dl (< 1.3 mmol/L) gen to glucose) and gluconeogenesis (formation of
Metabolic syndrome is common in Asian Indians in glucose from non-carbohydrates like proteins), which
Britain and in Africa. It is said that insulin resistance is contribute to hyperglycemia. Typical clinical features
common to all above risk factors and plays an etiological of hyperglycemia are polyuria, polydipsia, poly-
role. These persons have increased risk of developing phagia, weakness, weight loss, and blurring of vision.
Type 2 DM. • Glycosuria: Glycosuria results when blood glucose
level exceeds renal threshold (180 mg/dl or 10 mmol/
METABOLIC ALTERATIONS IN L in most individuals). Excess glucose increases
DIABETES MELLITUS osmolality of glomerular filtrate with resultant
In DM, there are abnormalities of carbohydrate osmotic diuresis and polyuria. This causes depletion
metabolism (hyperglycemia, glycosuria, impaired of water and electrolytes, cellular dehydration, and
glucose tolerance), protein metabolism (increased protein intense thirst (polydipsia). Insulin deficiency leads to
catabolism with muscle wasting, gluconeogenesis), and catabolism of proteins, and released amino acids are
fat metabolism (increased fatty acid synthesis, ketosis) used for formation of glucose (gluconeogenesis).
(Fig. 3.5). Metabolic alterations in type 2 DM are less Breakdown of lipids also occurs and coupled with
severe than in type 1 DM. proteolysis, lead to negative energy balance and
• Hyperglycemia: Hyperglycemia is due to deficient weight loss. This in turn induces polyphagia
uptake of glucose by muscle and fat cells (due to (increased appetite).

Fig. 3.5: Metabolic alterations in diabetes mellitus


44 Essentials of Clinical Pathology

• Ketosis: Insulin deficiency leads to increased


Box 3.3: Microangiopathy in diabetes mellitus
degradation of lipids (lipolysis), resulting in increased
levels of free fatty acids in circulation. These are • Risk is directly related to presence of high glucose
converted to acetoacetyl CoA in the liver. Acetoacetyl level for long duration
CoA, in turn, is converted to ketone bodies. If muscles • Improved glycemic control significantly reduces the
or other tissues do not utilize ketone bodies equal to risk
the rate of their formation, they accumulate in blood. • Consists of:
With the rise in blood levels of ketone bodies – Retinopathy: Visual loss can occur due to
(ketonemia), they are excreted in urine (ketonuria). vitreous hemorrhage (from proliferating retinal
Ketone bodies are strong acids, which dissociate and vessels) and maculopathy
– Nephropathy: Early stage is characterized by
release H+ ions. Bicarbonate removes these H+ ions
increased glomerular filtration rate and micro-
in plasma and the level of bicarbonate falls. This albuminuria; with progressive renal damage, overt
produces metabolic acidosis. Symptoms of metabolic proteinuria and renal failure develop.
acidosis are nausea, vomiting, and hyperpnea (air – Neuropathy: Postural hypotension, impotence,
hunger). Diabetic ketoacidosis (DKA) is a typical sensory and motor neuropathy, foot ulcer.
feature of type 1 DM.
• Hyperosmolar hyperglycemic state (HHS): This
occurs in type 2 patients due to combination of severe Average life expectancy of DM patients is reduced.
dehydration (secondary to sustained osmotic diuresis Usual causes of death in DM include myocardial
coupled with inadequate fluid intake) and hyper- infarction, stroke, renal failure, infections, and keto-
glycemia. It occurs usually in elderly with unrecog- acidotic or hyperosmolar hyperglycemic coma.
nized DM or DM of recent onset.
ROLE OF LABORATORY TESTS IN
LONG-TERM COMPLICATIONS OF DIABETES MELLITUS
DIABETES MELLITUS
In DM, applications of laboratory tests are as follows:
In long standing DM of both types, a wide variety of • Diagnosis of DM
lesions develop in many organs, which are important • Screening of DM
causes of morbidity and mortality. • Assessment of glycemic control
Macroangiopathy (Macrovascular disease): In DM, • Assessment of associated long-term risks
atherosclerosis of aorta and of medium size arteries (like • Management of acute metabolic complications.
coronary, cerebral, and peripheral) occurs earlier in life
Laboratory Tests for Diagnosis of
and is more extensive than in non-diabetic patients. It
Diabetes Mellitus
can cause myocardial infarction, cerebrovascular
accident, and gangrene of lower extremities. Patho- Diagnosis of DM is based exclusively on demonstration
genesis of atherosclerosis in DM is related to hyper- of raised blood glucose level (hyperglycemia).
insulinemia with peripheral insulin resistance and The current criteria (American Diabetes Association,
dyslipidemia (raised triglycerides, low high density 2004) for diagnosis of DM are as follows:
lipoprotein or HDL, and raised low density lipoprotein
Typical symptoms of DM (polyuria, polydipsia, weight
or LDL).
loss) and random plasma glucose ≥ 200 mg/dl (≥ 11.1
Microangiopathy (Microvascular disease): Micro- mmol/L)
angiopathy is due to poor diabetes control (Box 3.3). Or
Microangiopathy (thickening of walls of small blood Fasting plasma glucose ≥ 126 mg/dl (≥ 7.0 mmol/L)
vessels with narrowing of lumina) is common in kidneys Or
(leading to renal insufficiency), retina (visual impair- 2-hour post glucose load (75 g) value during oral
ment), and peripheral nerves (sensory, motor or glucose tolerance test ≥ 200 mg/dl (≥ 11.1 mmol/L).
autonomic neuropathy).
If any one of the above three criteria is present,
Infections: Diabetic patients are susceptible to infections confirmation by repeat testing on a subsequent day is
of skin, respiratory tract (pneumonia, tuberculosis), and necessary for establishing the diagnosis of DM. However,
kidneys (pyelonephritis). such confirmation by repeat testing is not required if
Diabetes Mellitus 45

patient presents with (a) hyperglycemia and ketoacidosis, Addition of sodium fluoride is not necessary if plasma
or (b) hyperosmolar hyperglycemia. is separated from whole blood within 1 hour of blood
The tests used for laboratory diagnosis of DM are (1) collection.
estimation of blood glucose and (2) oral glucose tolerance Plasma is preferred for estimation of glucose since
test. whole blood glucose is affected also by concentration of
proteins (especially hemoglobin).
Estimation of Blood Glucose There are various methods for estimation of blood
Measurement of blood glucose level is a simple test to glucose:
assess carbohydrate metabolism in DM (Fig. 3.6). Since • Chemical methods:
– Orthotoluidine method
glucose is rapidly metabolized in the body, measurement
– Blood glucose reduction methods using
of blood glucose is indicative of current state of
neocuproine, ferricyanide, or copper.
carbohydrate metabolism.
Glucose concentration can be estimated in whole Chemical methods are less specific but are cheaper as
blood (capillary or venous blood), plasma or serum. compared to enzymatic methods.
However, concentration of blood glucose differs • Enzymatic methods: These are specific for glucose.
according to nature of the blood specimen. Plasma – Glucose oxidase-peroxidase
glucose is about 15% higher than whole blood glucose – Hexokinase
(the figure is variable with hematocrit). During fasting – Glucose dehydrogenase
state, glucose levels in both capillary and venous blood Chemical methods have now been replaced by
enzymatic methods.
are about the same. However, postprandial or post
glucose load values are higher by 20-70 mg/dl in Terms used for blood glucose specimens: Depending
capillary blood than venous blood. This is because venous on the time of collection, different terms are used for
blood is on a return trip after delivering blood to the blood glucose specimens.
tissues. • Fasting blood glucose: Sample for blood glucose is
When whole blood is left at room temperature after withdrawn after an overnight fast (no caloric intake
collection, glycolysis reduces glucose level at the rate of for at least 8 hours).
about 7 mg/dl/hour. Glycolysis is further increased in • Post meal or postprandial blood glucose: Blood
sample for glucose estimation is collected 2 hours after
the presence of bacterial contamination or leucocytosis.
the subject has taken a normal meal.
Addition of sodium fluoride (2.5 mg/ml of blood)
• Random blood glucose: Blood sample is collected at
maintains stable glucose level by inhibiting glycolysis.
any time of the day, without attention to the time of
Sodium fluoride is commonly used along with an
last food intake.
anticoagulant such as potassium oxalate or EDTA.
Oral Glucose Tolerance Test (OGTT)
Glucose tolerance refers to the ability of the body to
metabolize glucose. In DM, this ability is impaired or
lost and glucose intolerance represents the fundamental
pathophysiological defect in DM. OGTT is a provocative
test to assess response to glucose challenge in an
individual (Fig. 3.7).
American Diabetes Association does not
recommend OGTT for routine diagnosis of type 1 or
type 2 DM. This is because fasting plasma glucose cut-
off value of 126 mg/dl identifies the same prevalence of
abnormal glucose metabolism in the population as
OGTT. World Health Organization (WHO) recommends
OGTT in those cases in which fasting plasma glucose is
in the range of impaired fasting glucose (i.e. 100-125 mg/
Fig. 3.6: Blood glucose values in normal individuals, dl). Both ADA and WHO recommend OGTT for
prediabetes, and diabetes mellitus diagnosis of gestational diabetes mellitus.
46 Essentials of Clinical Pathology

3. A single venous blood sample is collected 2 hours


after the glucose load. (Previously, blood samples
were collected at ½, 1, 1½, and 2 hours, which is no
longer recommended).
4. Plasma glucose is estimated in fasting and 2-hour
venous blood samples.
Interpretation of blood glucose levels is given in Table
3.2.
OGTT in gestational diabetes mellitus: Impairment of
glucose tolerance develops normally during pregnancy,
particularly in 2nd and 3rd trimesters. Following are the
recent guidelines of ADA for laboratory diagnosis of
GDM:
• Low-risk pregnant women need not be tested if all of
the following criteria are met, i.e. age below 25 years,
normal body weight (before pregnancy), absence of
Fig. 3.7: Oral glucose tolerance curve diabetes in first-degree relatives, member of an ethnic
group with low prevalence of DM, no history of poor
obstetric outcome, and no history of abnormal glucose
Preparation of the Patient tolerance.
• Patient should be put on a carbohydrate-rich, • Average-risk pregnant women (i.e. who are in
unrestricted diet for 3 days. This is because carbo- between low and high risk) should be tested at 24-28
hydrate-restricted diet reduces glucose tolerance. weeks of gestation.
• Patient should be ambulatory with normal physical • High-risk pregnant women i.e. those who meet any
activity. Absolute bed rest for a few days impairs one of the following criteria should be tested
glucose tolerance. immediately: marked obesity, strong family history
• Medications should be discontinued on the day of of DM, glycosuria, or personal history of GDM.
testing. Initially, fasting plasma glucose or random plasma
• Exercise, smoking, and tea or coffee are not allowed glucose should be obtained. If fasting plasma glucose is
during the test period. Patient should remain seated. ≥ 126 mg/dl or random plasma glucose is ≥ 200 mg/dl,
• OGTT is carried out in the morning after patient has repeat testing should be carried out on a subsequent day
fasted overnight for 8-14 hours. for confirmation of DM. If both the tests are normal, then
OGTT is indicated in average-risk and high-risk pregnant
Test women.
There are two approaches for laboratory diagnosis
1. A fasting venous blood sample is collected in the of GDM
morning. • One step approach
2. Patient ingests 75 g of anhydrous glucose in 250-300 • Two step approach
ml of water over 5 minutes. (For children, the dose is In one step approach, 100 gm of glucose is adminis-
1.75 g of glucose per kg of body weight up to tered to the patient and a 3-hour OGTT is performed.
maximum 75 g of glucose). Time of starting glucose This test may be cost-effective in high-risk pregnant
drink is taken as 0 hour. women.

Table 3.2: Interpretation of oral glucose tolerance test


Parameter Normal Impaired Impaired glucose Diabetes mellitus
fasting glucose tolerance

1. Fasting (8 hr) < 100 100-125 — ≥ 126


2. 2 hr OGTT < 140 <140 140-199 ≥ 200
Diabetes Mellitus 47

In two-step approach, an initial screening test is done Screening for type 1 DM: Type 1 DM is detected early after
in which patient drinks a 50 g glucose drink irrespective its onset since it has an acute presentation with
of time of last meal and a venous blood sample is characteristic clinical features. Therefore, it is not
collected 1 hour later (O’Sullivan’s test). GDM is excluded necessary to screen for type 1 DM by estimation of blood
if glucose level in venous plasma sample is below 140 glucose. Detection of immunologic markers (mentioned
mg/dl. If level exceeds 140 mg/dl, then the complete earlier) has not been recommended to identify persons
100 g, 3-hour OGTT is carried out. at risk.
In the 3-hour OGTT, blood samples are collected in
Screening for GDM: Given earlier under OGTT in
the morning (after 8-10 hours of overnight fasting), and
gestational diabetes mellitus.
after drinking 100 g glucose, at 1, 2, and 3 hours. For
diagnosis of GDM, glucose concentration should be
Laboratory Tests to Assess Glycemic Control
above the following cut-off values in 2 or more of the
venous plasma samples: There is a direct correlation between the degree of blood
• Fasting: 95 mg/dl glucose control in DM (both type 1 and type 2) and the
• 1 hour: 180 mg/dl development of microangiopathic complications i.e.
• 2 hour: 155 mg/dl nephropathy, retinopathy, and neuropathy. Maintenance
• 3 hour: 140 mg/dl of blood glucose level as close to normal as possible
(referred to as tight glycemic control) reduces the risk of
Laboratory Tests for Screening of microvascular complications. There is also association
Diabetes Mellitus between persistently high blood glucose values in DM
with increased cardiovascular mortality.
Aim of screening is to identify asymptomatic individuals
Following methods can monitor degree of glycemic
who are likely to have DM. Since early detection and
control:
prompt institution of treatment can reduce subsequent
• Periodic measurement of glycated hemoglobin (to
complications of DM, screening may be an appropriate
assess long-term control).
step in some situations.
• Daily self-assessment of blood glucose (to assess day-
Screening for type 2 DM: Type 2 DM is the most common to-day or immediate control).
type of DM and is usually asymptomatic in its initial
stages. Its onset occurs about 5-7 years before clinical Glycated Hemoglobin
diagnosis. Evidence indicates that complications of type (Glycosylated Hemoglobin, HbA1C)
2 DM begin many years before clinical diagnosis.
American Diabetes Association recommends screening Glycated hemoglobin refers to hemoglobin to which
for type 2 DM in all asymptomatic individuals ≥ 45 years glucose is attached nonenzymatically and irreversibly;
of age using fasting plasma glucose. If fasting plasma its amount depends upon blood glucose level and
glucose is normal (i.e. < 100 mg/dl), screening test should lifespan of red cells.
be repeated every three years.
Another approach is selective screening i.e. screening Hemoglobin + Glucose ↔ Aldimine → Glycated hemoglobin
individuals at high risk of developing type 2 DM i.e. if
one or more of the following risk factors are present- Plasma glucose readily moves across the red cell
obesity (body mass index ≥ 25.0 kg/m2), family history membranes and is being continuously combined with
of DM (first degree relative with DM), high-risk ethnic hemoglobin during the lifespan of the red cells (120 days).
group, hypertension, dyslipidemia, impaired fasting Therefore, some hemoglobin in red cells is present
glucose, impaired glucose tolerance, or history of GDM. normally in glycated form. Amount of glycated
In such cases, screening is performed at an earlier age hemoglobin in blood depends on blood glucose
(30 years) and repeated more frequently. concentration and lifespan of red cells. If blood glucose
Recommended screening test for type 2 DM is fasting concentration is high, more hemoglobin is glycated. Once
plasma glucose. If ≥126 mg/dl, it should be repeated on formed, glycated hemoglobin is irreversible. Level of
a subsequent day for confirmation of diagnosis. If <126 glycated hemoglobin is proportional to the average
mg/dl, OGTT is indicated if clinical suspicion is strong. glucose level over preceding 6-8 weeks (about 2 months).
A 2-hour post-glucose load value in OGTT ≥200 mg/dl Glycated hemoglobin is expressed as a percentage of total
is indicative of DM and should be repeated on a different hemoglobin. Normally, less than 5% of hemoglobin is
day for confirmation. glycated.
48 Essentials of Clinical Pathology

Self-Monitoring of Blood Glucose (SMBG)


Numerous prospective studies have demonstrated
that a good control of blood glucose reduces the Diabetic patients are taught how to regularly monitor
development and progression of microvascular their own blood glucose levels. Regular use of SMBG
complications (retinopathy, nephropathy, and
devices (portable glucose meters) by diabetic patients has
peripheral neuropathy) of diabetes mellitus. Mean
improved the management of DM. With SMBG devices,
glycated hemoglobin level correlates with the risk of
these complications. blood glucose level can be monitored on day-to-day basis
and kept as close to normal as possible by adjusting
The terms glycated hemoglobin, glycosylated insulin dosage. SMBG devices measure capillary whole
hemoglobin, glycohemoglobin, HbA1, and HbA1c are blood glucose obtained by fingerprick and use test strips
often used interchangeably in practice. Although these that incorporate glucose oxidase or hexokinase. In some
terms refer to hemoglobins that contain nonenzy- strips, a layer is incorporated to exclude blood cells so
matically added glucose residues, hemoglobins thus
that glucose in plasma is measured. Aim of achieving
modified differ. Most of the studies have been carried
out with HbA1c. tight glycemic control introduces the risk of severe
Glycated hemoglobin should be routinely measured hypoglycemia. Daily use of SMBG devices can avoid
in all diabetic patients (both type 1 and type 2) at regular major hypoglycemic episodes.
intervals to assess degree of long-term glycemic control. SMBG devices yield unreliable results at very high
Apart from mean glycemia (over preceding 120 days), and very low glucose levels. It is necessary to periodically
glycated hemoglobin level also correlates with the risk check the performance of the glucometer by measuring
of the development of chronic complications of DM.
parallel venous plasma glucose in the laboratory.
In DM, it is recommended to maintain glycated
Portable glucose meters are used by patients for day-
hemoglobin level to less than 7%.
Spurious results of glycated hemoglobin are seen in to-day self-monitoring, by physicians in their OPD
reduced red cell survival (hemolysis), blood loss, and clinics, and by health care workers for monitoring
hemoglobinopathies. admitted patients at the bedside. These devices should
In DM, if glycated hemoglobin is less than 7%, it not be used for diagnosis and population screening of
should be measured every 6 months. If >8%, then more DM as they lack precision and there is variability of
frequent measurements (every 3 months) along with results between different meters.
change in treatment are advocated.
There are various methods for measurement of Goal of tight glycemic control in type 1 DM patients
glycated hemoglobin such as chromatography, immuno- on insulin can be achieved through self-monitoring of
assay, and agar gel electrophoresis. blood glucose by portable blood glucose meters.
Role of glycated hemoglobin in management of DM
is highlighted in Box 3.4.
Glycosuria

Box 3.4: Glycated hemoglobin Semiquantitative urine glucose testing for monitoring
of diabetes mellitus in home setting is not recommen-
• Hemoglobin A1C of 6% corresponds to mean serum glucose ded. This is because (1) even if glucose is absent in
level of 135 mg/dl. With every rise of 1%, serum glucose
increases by 35 mg/dl. Approximations are as follows:
urine, no information about blood glucose concen-
– Hb A1C 7%: 170 mg/dl tration below the renal threshold (which itself is
– Hb A1C 8%: 205 mg/dl variable) is obtained (Normally, renal threshold is
– Hb A1C 9%: 240 mg/dl around 180 mg/dl; it tends to be lower in pregnancy (140
– Hb A1C 10%: 275 mg/dl mg/dl) and higher in old age and in long-standing
– Hb A1C 11%: 310 mg/dl
– Hb A1C 12%: 345 mg/dl
diabetics; in some normal persons it is low), (2) urinary
• Assesses long-term control of DM (thus indirectly glucose testing cannot detect hypoglycemia, and (3)
confirming plasma glucose results or self-testing results). concentration of glucose in urine is affected by urinary
• Assesses whether treatment plan is working concentration. Semiquantitative urine glucose testing
• Measurement of glycated hemoglobin does not replace for monitoring has now been replaced by self-testing
measurement of day-to-day control by glucometer devices. by portable glucose meters.
Diabetes Mellitus 49

Laboratory Tests to Assess Long-term Risks Albumin excretion rate is intermediate between normal
(normal albumin excretion in urine is < 30 mg/24 hours)
Urinary Albumin Excretion and overt albuminuria (> 300 mg/24 hours). Significance
of microalbuminuria in DM is as follows:
Diabetes mellitus is one of the leading causes of renal
• It is the earliest marker of diabetic nephropathy.
failure. Diabetic nephropathy develops in around 20-30%
Early diabetic nephropathy is reversible.
of patients with type 1 or type 2 DM. Diabetic nephro-
• It is a risk factor for cardiovascular disease in both
pathy progresses through different stages as shown in type 1 and type 2 patients.
Figure 3.8. Hypertension also develops along the course • It is associated with higher blood pressure and poor
of nephropathy with increasing albumin excretion. glycemic control.
Evidence indicates that if diabetic nephropathy is Specific therapeutic interventions such as tight
detected early and specific treatment is instituted, further glycemic control, administration of ACE (angiotensin-
progression of nephropathy can be significantly converting enzyme) inhibitors, and aggressive treatment
ameliorated. Early detection of diabetic nephropathy is of hypertension significantly slow down the progression
based on estimation of urinary albumin excretion. In all of diabetic nephropathy.
adult patients with DM, usual reagent strip test for In type 2 DM, screening for microalbuminuria
proteinuria should be carried out periodically. Positive should begin at the time of diagnosis, whereas in type
test means presence of overt proteinuria or clinical 1 DM, it should begin 5 years after diagnosis. At this
proteinuria and may be indicative of overt nephropathy. time, a routine reagent strip test for proteinuria is carried
In all such patients quantitation of albuminuria should out; if negative, testing for microalbuminuria is done.
be carried out to plan appropriate therapy. If the routine Thereafter, in all patients who test negative, screening
dipstick test for proteinuria is negative, test for for microalbuminuria should be repeated every year.
microalbuminuria should be carried out. Screening tests for microalbuminuria include:
The term ‘microalbuminuria’ refers to the urinary • Albumin to creatinine ratio in a random urine sample
excretion of albumin below the level of detection by • Urinary albumin excretion in a 24-hour urine sample.
routine dipstick testing but above normal (30-300 mg/ Reagent strip tests to detect microalbuminuria are
24 hrs, 20-200 μg/min, or 30-300 μg/mg of creatinine). available. Positive results should be confirmed by more

Fig. 3.8: Evolution of diabetic nephropathy. In 80% of patients with type 1 DM, microalbuminuria progresses in 10-15 years
to overt nephropathy that is then followed in majority of cases by progressive fall in GFR and ultimately end-stage renal
disease. Amongst patients with type 2 DM and microalbuminuria, 20-40% of patients progress to overt nephropathy, and
about 20% of patients with overt nephropathy develop end-stage renal disease. Abbreviation: GFR: Glomerular filtration rate
50 Essentials of Clinical Pathology

specific quantitative tests like radioimmunoassay and Laboratory Tests in the Management of Acute
enzyme immunoassay. For diagnosis of microalbumi- Metabolic Complications of Diabetes Mellitus
nuria, tests should be positive in at least two out of three The three most serious acute metabolic complications of
different samples over a 3 to 6 month period. DM are:
• Diabetic ketoacidosis (DKA)
• Hyperosmolar hyperglycemic state (HHS)
Lipid Profile • Hypoglycemia
The typical features of DKA are hyperglycemia,
Abnormalities of lipids are associated with increased risk
ketosis, and acidosis. The common causes of DKA are
of coronary artery disease (CAD) in patients with DM. infection, noncompliance with insulin therapy, alcohol
This risk can be reduced by intensive treatment of lipid abuse and myocardial infarction. Patients with DKA
abnormalities. Lipid parameters which should be present with rapid onset of polyuria, polydipsia,
measured include: polyphagia, weakness, vomiting, and sometimes
abdominal pain. Signs include Kussmaul’s respiration,
• Total cholesterol odour of acetone on breath (fruity), mental clouding, and
• Triglycerides dehydration. Classically, DKA occurs in type 1, while
• Low-density lipoprotein (LDL) cholesterol HHS is more typical of type 2 DM. However, both
• High-density lipoprotein (HDL) cholesterol complications can occur in either types. If untreated, both
events can lead to coma and death.
The usual pattern of lipid abnormalities in type 2
Hyperosmolar hyperglycemic state is characterized by
DM is elevated triglycerides, decreased HDL choleste- very high blood glucose level (> 600 mg/dl), hyper-
rol and higher proportion of small, dense LDL particles. osmolality (>320 mOsmol/kg of water), dehydration, lack
Patients with DM are categorized into high, intermediate of ketoacidosis, and altered mental status. It usually occurs
and low-risk categories depending on lipid levels in in elderly type 2 diabetics. Insulin secretion is adequate to
prevent ketosis but not hyperglycemia. Causes of HHS are
blood (Table 3.3).
illness, dehydration, surgery, and glucocorticoid therapy.
Annual lipid profile is indicated in all adult patients Differences between DKA and HHS are presented in
with DM. Table 3.4.
Table 3.3: Categorization of cardiovascular risk in diabetes mellitus
according to lipid levels (American Diabetes Association)
Category Low density lipoproteins High density lipoproteins Triglycerides

High-risk ≥130 < 35 (men) ≥ 400


< 45 (women)
Intermediate risk 100-129 35-45 200-399
Low-risk < 100 > 45 (men) < 200
> 55 (women)

Table 3.4: Comparison of diabetic ketoacidosis and hyperosmolar hyperglycemic state


Parameter Diabetic ketoacidosis Hyperosmolar hyperglycemic state

1. Type of DM in which more common Type 1 Type 2


2. Age Younger age Older age
3. Prodromal clinical features < 24 hrs Several days
4. Abdominal pain, Kussmaul’s respiration Yes No
5. Acidosis Moderate/Severe Absent
6. Plasma glucose > 250 mg/dl Very high (>600 mg/dl)
7. Serum bicarbonate <15 mEq/L >15 mEq/L
8. Blood/urine ketones ++++ ±
9. β-hydroxybutyrate High Normal or raised
10. Arterial blood pH Low (<7.30) Normal (>7.30)
11. Effective serum osmolality* Variable Increased (>320)
12. Anion gap** >12 Variable
*Osmolality: Number of dissolved (solute) particles in solution; normal: 275-295 mOsmol/kg
** Anion gap: Difference between sodium and sum of chloride and bicarbonate in plasma; normal average value is 12
Diabetes Mellitus 51

Laboratory evaluation consists of following investiga- Serum Osmolality can also be calculated by the
tions: following formula recommended by American Diabetes
• Blood and urine glucose Association:
• Blood and urine ketone
Effective serum osmolality (mOsm/kg) =
• Arterial pH, Blood gases
(2 × sodium mEq/L) + Plasma glucose (mg/dl)
• Serum electrolytes (sodium, potassium, chloride,
bicarbonate) 18
• Blood osmolality • Anion gap:
• Serum creatinine and blood urea. – Na+ – (Cl– + HCO3–): 8-16 mmol/L (Average 12)
– (Na + + K + ) – (Cl – + HCO3 – ): 10-20 mmol/L
Testing for ketone bodies: Ketone bodies are formed (Average 16)
from metabolism of free fatty acids and include • Serum sodium: 135-145 mEq/L
acetoacetic acid, acetone and β-hydroxybutyric acid. • Serum potassium: 3.5-5.0 mEq/L
Indications for testing for ketone bodies in DM include: • Serum chloride: 100-108 mEq/L
• At diagnosis of diabetes mellitus • Serum bicarbonate: 24-30 mEq/L
• At regular intervals in all known cases of diabetes,
during pregnancy with pre-existing diabetes, and in CRITICAL VALUES
gestational diabetes • Venous plasma glucose: > 450 mg/dl
• In known diabetic patients: during acute illness, • Strongly positive test for glucose and ketones in
persistent hyperglycemia (> 300 mgs/dl), pregnancy, urine
and clinical evidence of diabetic acidosis (nausea, • Arterial pH: < 7.2 or > 7.6
vomiting, abdominal pain). • Serum sodium: < 120 mEq/L or > 160 mEq/L
An increased amount of ketone bodies in patients • Serum potassium: < 2.8 mEq/L or > 6.2 mEq/L
with DM indicate impending or established diabetic • Serum bicarbonate: < 10 mEq/L or > 40 mEq/L
ketoacidosis and is a medical emergency. Method based • Serum chloride: < 80 mEq/L or > 115 mEq/L
on colorimetric reaction between ketone bodies and
BIBLIOGRAPHY
nitroprusside (by dipstick or tablet) is used for detection
of both blood and urine ketones. 1. American Diabetes Association. Diagnosis and
classification of diabetes mellitus. Diabetes Care 2004;
Test for urine ketones alone should not be used for
24:S5-S10.
diagnosis and monitoring of diabetic ketoacidosis. It is 2. American Diabetes Association. Gestational diabetes
recommended to measure β-hydroxybutyric acid (which mellitus. Diabetes Care 2004;27:S88-S90.
accounts for 75% of all ketones in ketoacidosis) for 3. American Diabetes Association. Hyperglycemic crises
diagnosis and monitoring DKA. in diabetes. Diabetes Care 2004;27:S94-S102.
4. American Diabetes Association. Screening for type 2
diabetes. Diabetes Care 2004;27:S11-S14.
REFERENCE RANGES 5. American Diabetes Association. Tests of glycemia in
• Venous plasma glucose: diabetes. Diabetes Care 2004;27:S91-S93.
6. Lernmark A. Type 1 diabetes. Clin Chem 1999;45:
Fasting: 60-100 mg/dl
1331-38.
At 2 hours in OGTT (75 gm glucose): <140 mg/dl 7. Lebovitz HE. Type 2 diabetes: An overview. Clin Chem
• Glycated hemoglobin: 4-6% of total hemoglobin 1999;45:1339-45.
• Lipid profile: 8. Reaven GM. The metabolic syndrome: Requiescat in
– Serum cholesterol: Desirable level: <200 mg/dl pace. Clin Chem 2005;51:931-8.
9. Reinauer H, Home PD, Kanagasabapathy AS, Heuck
– Serum triglycerides: Desirable level: <150 mg/dl
C. Laboratory diagnosis and monitoring of diabetes
– HDL cholesterol: ≥60 mg/dl mellitus. Geneva. World Health Organization, 2002.
– LDL cholesterol: <130 mg/dl 10. Sacks DB, Bruns DE, Goldstein DE, Maclaren NK,
– LDL/HDL ratio: 0.5-3.0 McDonald JM, Parrott M. Guidelines and recommen-
• C-peptide: 0.78-1.89 ng/ml dations for laboratory analysis in the diagnosis and
management of diabetes mellitus. Clin Chem 2002;
• Arterial pH: 7.35-7.45 48:436-72.
• Serum or plasma osmolality: 275-295 mOsm/kg of 11. Trachtenbarg DE. Diabetic ketoacidosis. Am Fam
water. Physician 2005;71:1705-14.
52 Essentials of Clinical Pathology
4

Liver Function Tests

FUNCTIONS OF LIVER
1. Excretory function: Liver cells metabolize and excrete
endogenous as well as exogenous substances. Liver
regulates bilirubin metabolism by secretion and
excretion of bilirubin.
2. Synthetic function: Synthesis of proteins like
albumin, α- and β-globulins, transport proteins and
many coagulation proteins occurs in the liver. Liver
also produces triglycerides, cholesterol, lipoproteins,
and primary bile acids. Albumin maintains osmotic
pressure of plasma, transports various compounds,
and acts as a protein reserve. Liver does not
synthesize immunoglobulins and complement.
3. General metabolic functions: Liver regulates carbo-
hydrate, lipid, and protein metabolism.
Glycogen, derived from monosaccharides, is
stored in the liver. When carbohydrate intake is
reduced, blood glucose level is maintained by
breakdown of stored glycogen (glycogenolysis). If
needed, amino acids and fat are converted to glucose
by the liver (gluconeogenesis).
Synthesis of triglycerides, phospholipids, choles-
terol, and lipoproteins occurs in the liver. Liver also
esterifies cholesterol, and forms bile acids from
cholesterol (Fig. 4.1). Bile acids are essential for fat Fig. 4.1: Formation of bile acids and bile salts
absorption from the intestine. Lipoproteins help in
transport of fats.
Besides synthesis of various proteins and NORMAL BILIRUBIN METABOLISM
enzymes, liver is the site for deamination and
transamination of amino acids. Ammonia is Bilirubin is mostly (85%) produced from breakdown
converted to urea in the urea cycle and detoxified in of hemoglobin of old red cells in reticuloendothelial
the liver. cells (macrophages), mainly in spleen. A smaller
4. Liver is the storage site for iron, glycogen, and amount is derived from premature destruction of red cell
vitamins. precursors in bone marrow, and from myoglobin,
5. During fetal life, hematopoiesis occurs in the liver. It
cytochromes, and peroxidases. Steps in metabolism of
is also a site for destruction of damaged red cells
(immune hemolysis). bilirubin are outlined below (Fig. 4.2).
6. Liver is the major organ for catabolism of steroid 1. Hemoglobin is degraded within macrophages to form
hormones. heme and globin; globin consists of amino acids
Liver Function Tests 53

Fig. 4.2: Normal bilirubin metabolism

which are recycled. Heme (iron + protoporphyrin) are necessary for digestion and absorption of fat from
releases iron, which is stored as ferritin. the small intestine.
Protoporphyrin is first converted to biliverdin, which 6. When bilirubin reaches the large intestine, it is
is then reduced to bilirubin. converted by bacterial action to a group of
2. Bilirubin is released from macrophages into the compounds known as urobilinogen by the action of
circulation where it binds with albumin. This is called bacterial enzymes.
as unconjugated bilirubin. It is soluble in lipid but 7. Most of the urobilinogen is excreted in feces as
insoluble in water. urobilin and is responsible for brown coloration of
3. Bilirubin-albumin complex reaches the liver where feces. A part of urobilinogen is absorbed into the
it is taken up by the hepatocytes. Bilirubin is set free circulation from where it reaches the liver, is taken
in the cytoplasm, while albumin is released back into by the hepatocytes, and is again re-excreted in bile
the circulation. (enterohepatic circulation). A small amount of
4. Bilirubin is conjugated with glucuronic acid to form urobilinogen in circulation escapes clearance by the
bilirubin monoglucuronide and diglucuronide liver and is excreted in urine.
(conjugated bilirubin); this process is mediated by the
enzyme glucuronyl transferase. Conjugated bilirubin INDICATIONS AND LIMITATIONS OF
is more soluble in water. LIVER FUNCTION TESTS
5. Conjugated bilirubin is secreted from the hepatocyte
into the biliary canaliculi, from where it passes into Liver function tests (LFT) are the various laboratory tests
the bile duct and gallbladder along with bile (bilirubin that are used to:
monoglucuronide 25% and bilirubin diglucuronide • Screen for liver disease;
75%). Bilirubin reaches the small intestine via the • Identify the nature of liver disease (hepatocellular,
common bile duct. Bile also contains bile salts, which cholestatic, or infiltrative);
54 Essentials of Clinical Pathology

• Assess severity and prognosis of liver disease; and Table 4.1: Commonly performed liver function tests
• Follow up the course of liver disease
Test Hepatic cause of abnormality
Box 4.1: Limitations of liver function tests
1. Serum alanine Hepatocellular injury
• Do not necessarily assess liver function aminotransferase
• Lack sensitivity (i.e. may be normal in some liver
2. Serum aspartate Hepatocellular injury
diseases like cirrhosis)
aminotransferase
• Lack specificity (i.e. may be abnormal in non-liver
disorders e.g. serum albumin is low in nephrotic 3. Serum alkaline Cholestasis
syndrome and in cirrhosis) phosphatase
4. Serum bilirubin Defective conjugation or excretion
5. Serum albumin Decreased synthesis
The term ‘liver function tests’ is a misnomer since
most of these tests are used for identification of liver CLASSIFICATION OF
disease, its severity, and its type and do not necessarily
LIVER FUNCTION TESTS
assess liver function. Generally these tests, which are
performed in combination, are abnormal in liver disease, Liver function tests can be classified as follows:
and the pattern of abnormality is indicative of the nature 1. Tests that assess excretory function of the liver:
of liver disease. Since liver has a large amount of Bilirubin in serum and urine, and urobilinogen in
urine and feces.
anatomical and functional reserve and capacity for rapid
2. Tests that assess synthetic and metabolic functions
regeneration, functional deficiency becomes apparent if
of the liver: Serum proteins, serum albumin, serum
there is an extensive liver damage. Also, these tests are albumin/globulin (A/G) ratio, prothrombin time
not specific for liver disease (except serum bile acids) and (PT), and blood ammonia level.
are abnormal in various non-hepatic conditions (Boxes 3. Tests that assess hepatic injury (liver enzyme
4.1 and 4.2). Therefore, laboratory tests should be studies): Serum alanine aminotransferase (ALT),
selected and interpreted in the context of clinical serum aspartate aminotransferase (AST), serum
features and other investigations. An isolated alkaline phosphatase, serum γ-glutamyl transferase
abnormality of a single liver function test usually means (GGT), and 5’-nucleotidase (5’-NT).
a non-hepatic cause. If several liver function tests are 4. Tests that assess clearance of exogenous substances
simultaneously abnormal, then hepatic etiology is likely. by the liver: Bromosulphthalein excretion test.
Commonly performed liver function tests are listed
in Table 4.1.
Box 4.2: Non-hepatic causes of abnormal liver Tests that Assess Excretory
function tests Function of the Liver
• Increased serum bilirubin: Jaundice
– Hemolysis
– Ineffective erythropoiesis
Jaundice (from French jaune, meaning yellow) or icterus
– Resorption of a large hematoma refers to yellow discoloration of skin, sclera, and mucous
membranes due to increased level of serum bilirubin.
• Increased aminotransferases:
Jaundice becomes clinically evident when serum
– Muscle injury
– Alcohol abuse
bilirubin level exceeds 2.0 mg/dl.
– Myocardial infarction There are various methods for classification of
jaundice as follows:
• Increased serum alkaline phosphatase:
1. According to the main type of bilirubin increased
– Pregnancy
– Bone disease
in plasma:
• Predominantly unconjugated hyperbilirubinemia:
• Low serum albumin: Indirect or unconjugated bilirubin is > 85% of total;
– Poor nutritional status
causes are hemolysis, resorption of a large
– Proteinuria
– Malabsorption
hematoma, ineffective erythropoiesis, Gilbert’s
– Severe illness causing protein catabolism syndrome, physiologic jaundice of newborn, and
Crigler-Najjar syndrome.
Liver Function Tests 55

• Predominantly conjugated hyperbilirubinemia: and feces. Jaundice is usually mild (serum bilirubin
Direct or conjugated bilirubin is >50% of total; <5.0 mg/dl; conjugated bilirubin is <15% of total).
causes are hepatitis, cirrhosis, cholestasis, drugs The cause can usually be identified by examination
(anabolic steroids, oral contraceptives), toxins, of a stained blood smear and hematological
Dubin-Johnson syndrome, and Rotor syndrome. investigations. In jaundiced newborns, rapidly rising
• Mixed (conjugated + unconjugated) hyperbili- unconjugated bilirubin needs careful management to
rubinemia: Conjugated bilirubin is 20-50% of total; prevent kernicterus.
it results from viral or alcoholic hepatitis. 2. Hepatic jaundice: In hepatic disease, unconjugated,
2. According to etiology: conjugated, or both are increased.
• Hemolytic: The increased rate of red cell destruc- a. Unconjugated hyperbilirubinemia:
tion causes increased haemoglobin breakdown to 1. Defective uptake of bilirubin by liver cells from
bilirubin in reticuloendothelial cells; this exceeds blood: Gilbert’s syndrome is the most common
the capacity of conjugation in liver. cause of unconjugated hyperbilirubinemia,
• Hepatocellular: Inability of hepatocytes to conju- affecting 5% of general population. It is a
gate and/or excrete bilirubin. familial, benign disease with autosomal domi-
• Obstructive: Failure of excretion of conjugated nant mode of inheritance. Raised bilirubin is
bilirubin into the intestine, causing its usually noticed during routine laboratory
regurgitation in circulation.
examination. There is defective uptake by
3. According to site of disease:
hepatocytes and mild deficiency of glucuronyl
• Prehepatic
transferase. Jaundice is mild and fluctuating,
• Hepatic
and may go unnoticed for years. Jaundice
• Posthepatic
becomes noticeable during illness or after
A simple classification is division of jaundice into
fasting. Mildly elevated serum bilirubin is the
three main types: prehepatic, hepatic, and
sole abnormality; other LFTs are normal.
posthepatic. This classification is the basis for
identifying the cause of jaundice (Table 4.2). 2. Defective conjugation of bilirubin:
1. Prehepatic jaundice: There is excessive formation of Crigler-Najjar syndrome: This is of two types.
bilirubin exceeding the capacity of the liver to Type I is characterized by autosomal recessive
conjugate it for excretion. The type of bilirubin mode of inheritance, complete absence of
increased in serum is of unconjugated type. Bilirubin glucuronyl transferase, severe unconjugated
is absent in urine since unconjugated bilirubin is hyperbilirubinemia, and kernicterus (deposition
water-insoluble. Urobilinogen is increased in urine of bilirubin in basal ganglia of brain). Type II is a

Table 4.2: Classification of jaundice according to the site of disease

Prehepatic jaundice
• Hemolytic anemia
• Ineffective erythropoiesis (megaloblastic anemia, thalassemias)
• Resorption of a large hematoma
Hepatic jaundice
• Predominantly unconjugated hyperbilirubinemia
– Gilbert’s syndrome
– Crigler-Najjar syndrome
– Physiologic jaundice of newborn
• Predominantly conjugated hyperbilirubinemia
– Hepatocellular diseases: viral hepatitis, toxic hepatitis, alcoholic hepatitis, active cirrhosis
– Intrahepatic cholestasis: Dubin-Johnson syndrome, drugs, primary biliary cirrhosis, primary sclerosing
cholangitis, biliary atresia
Posthepatic jaundice
• Carcinoma of head of pancreas
• Carcinoma of ampulla of Vater
• Secondaries in porta hepatis
• Gallstones in or stricture of common bile duct
56 Essentials of Clinical Pathology

less severe disease in which some amount of mation and destruction of both intrahepatic
enzyme activity is present. and extra-hepatic bile ducts. Associated
Physiologic jaundice of newborn: This is a tran- inflammatory bowel disease is often present.
sient increase of unconjugated bilirubin which is Serum alkaline phosphatase is elevated and
observed in almost all newborns. It usually many patients have circulating perinuclear
develops during the 2nd to 4th day after birth antineutrophil cytoplasmic antibodies.
with return to normal bilirubin level by 7th to 3. Posthepatic jaundice: This is also called as
10th day. It is because of deficiency of glucuronyl obstructive jaundice, surgical jaundice, or
transferase leading to impaired conjugation extrahepatic cholestasis.
during the first few days of life. Different sites of cholestasis and causative
b. Conjugated hyperbilirubinemia: disorders are shown in Figure 4.3.
1. Hepatocellular disease: Liver enzymes (aspar- Obstruction of extrahepatic biliary tract
tate aminotransferase and alanine amino- prevents flow of bile into the duodenum. This
transferase) are markedly elevated, and serum causes “regurgitation” of conjugated bilirubin
bilirubin is usually in the range of 4.0 to 8.0 into the circulation. (Biliary canaliculi distend
mg/dl. Conjugated bilirubin is 20-50% of total and rupture due to backpressure of bile and
bilirubin. In hepatocellular injury, both conju- conjugated bilirubin escapes into the
gated and unconjugated bilirubins are increa- sinusoids). Conjugated bilirubin is usually
sed. Unconjugated bilirubin is increased due >50% of total in posthepatic jaundice. Urinary
to reduced ability of liver cells to conjugate and fecal urobilinogen are decreased, faeces are
bilirubin. Conjugated bilirubin is raised from clay-colored, and bilirubin (being conjugated
cholestasis due to hepatocyte swelling. and water-soluble) appears in urine.
2. Intrahepatic cholestasis: In intrahepatic choles- Differences between three main types of jaundice are
tasis, there may be (1) impairment of secretion given in Table 4.3.
of bilirubin from hepatocytes into the biliary Investigation of a case of jaundice is shown in Figure 4.4.
canaliculi; (2) obstruction of bile flow in The tests employed to assess excretory function of
canaliculi by swollen hepatocytes;or (3) liver are serum/urine bilirubin and urine/fecal
damage to intrahepatic canaliculi. urobilinogen.
In Dubin-Johnson syndrome, there is a Serum Bilirubin
defect in the excretion of bilirubin by hepato-
Normal serum bilirubin is less than 1 mg/dl. There are two
cytes, and liver is darkly pigmented due to
types of bilirubin in plasma: unconjugated (indirect-
accumulation of polymerized epinephrine
reacting) and conjugated (direct-reacting) (Box 4.3).
metabolites. Bromosulphthalein excretion test
Normally, conjugated bilirubin is 10% or less, while
is abnormal. In Rotor syndrome, there is impai-
red excretion of bilirubin but without pigmen-
tation of liver; bromosulphthalein test is
normal. Alkaline phosphatase is normal in both
conditions.
Drugs commonly associated with choles-
tatic injury are oral contraceptives, anabolic
steroids, oral anti-diabetics, phenothiazines,
and erythromycin.
In primary biliary cirrhosis, there is auto-
immune destruction of intrahepatic bile ducts.
It predominantly occurs in middle-aged
females and is characterized by chronic eleva-
tion of alkaline phosphatase and positive anti-
mitochondrial antibody in serum. There is an
association with other autoimmune disorders.
Primary sclerosing cholangitis is an auto-
immune disorder occurring in young to
middle-aged men in whom there is inflam- Fig. 4.3: Sites of cholestasis
Liver Function Tests 57

Table 4.3: Differential diagnosis of three main types of jaundice

Parameter Prehepatic jaundice Hepatocellular Posthepatic


jaundice jaundice

1. Basic mechanism of Hemolysis leading to excess Deficient uptake, Deficient excretion due to
raised bilirubin production conjugation, or obstruction of biliary tract
excretion by hepato-
cytes
2. Type of serum bilirubin Mainly unconjugated Unconjugated + Mainly conjugated (>50%)
increased Conjugated
3. Urine bilirubin Absent Present Present
4. Urine urobilinogen Increased Variable Decreased
5. Prototype Hemolytic anemia Viral hepatitis Common duct stone
6. Prothrombin time Normal Abnormal that is not Abnormal that is corrected
corrected with with vit K
vitamin K
7. Additional features Features of haemolysis on Marked rise of serum Marked rise of serum ALP
blood smear (reticulocytosis, ALT and AST (>3 times normal)
low haptoglobin, low hemoglobin)

ALT: Alanine aminotransferase; AST: Aspartate aminotransferase; ALP: Alkaline phosphatase

Fig. 4.4: Investigation of jaundice. ALT: alanine aminotransferase; AST: aspartate aminotransferase;
ALP: alkaline phosphatase

unconjugated bilirubin is 90% or more. This is because very small amount. In cholestasis, proportion of δ-bili-
conjugated bilirubin is rapidly secreted into the bile after rubin increases. Owing to its longer half-life, it is cleared
its formation and removed through the gut. Conjugated slowly from circulation. Conjugated bilirubin is weakly
bilirubin is composed of blirubin glucuronide, bilirubin bound to albumin, is water-soluble, and can be excreted
diglucuronide, and delta (δ) bilirubin. Delta bilirubin in urine. Unconjugated bilirubin is tightly bound to
represents bilirubin covalently bound to albumin in albumin and is water-insoluble. As bilirubin is altered by
circulation. Normally, δ bilirubin is absent or present in exposure to light, sample should be kept in the dark.
58 Essentials of Clinical Pathology

Fig. 4.5: Estimation of serum bilirubin by diazo method

There are two methods for estimation of serum also contain carotene and other pigments, which absorb
bilirubin: diazo method and spectrophotometry. light at the same wavelength. In newborns, other
In diazo method, total and direct-reacting bilirubin pigments are absent.
are measured, and indirect bilirubin is obtained by
subtracting direct from total bilirubin (Fig. 4.5). Urine Bilirubin and Urobilinogen
Estimation of both types of bilirubin is helpful in the See Chapter 1 “Examination of Urine”.
differential diagnosis of jaundice. In post-hepatic type
of jaundice, direct bilirubin is the predominant form (> Tests which Assess Synthetic and Metabolic
50% of the total). In hepatocellular jaundice, direct Functions of Liver
bilirubin is usually between 20-50% of total. Indirect Markers of hepatic synthetic function are serum albumin
bilirubin predominates in hemolysis, Gilbert’s syndrome, and prothrombin time. Hypoalbuminemia and a pro-
and Crigler-Najjar syndrome (direct bilirubin is < 15% longed prothrombin time indicate severe functional
of total). impairment of liver.
Proteins
Box 4.3: Serum bilirubin
Liver is the sole site for the synthesis of most of the plasma
• Direct bilirubin (Conjugated bilirubin): It reacts
proteins, except gamma globulins which are synthesized
directly with diazo reagent. It consists of mono-
conjugated bilirubin, diconjugated bilirubin, and bilirubin
by plasma cells. Concentration of total serum proteins
tightly bound to albumin (delta bilirubin). in adults is about 5.5 to 8.0 gm/dl, while that of serum
albumin is 3.5 to 5.0 gm/dl. Serum albumin comprises
• Indirect bilirubin (Unconjugated bilirubin): It reacts
about 60% of total serum proteins.
with diazo reagent in the presence of alcohol. It consists
of bilirubin bound to albumin. It is calculated as ‘total Tests for proteins in liver disease include total
bilirubin minus direct bilirubin’. serum proteins, serum albumin, calculation of serum
albumin/globulin ratio (normal ratio is >1.5), and serum
protein electrophoresis.
Direct spectroscopic estimation is used for measurement 1. Total serum proteins: These can be estimated by
of total serum bilirubin in newborns and infants (<3 refractometer method or by biuret method.
months of age). Concentration of serum bilirubin is In refractometer method, refractive index of the
directly proportional to its absorbance in a solution (which depends on solute concentration) is
spectrophotometer at 454 nm. This method cannot be measured. Refractive index varies mainly with concen-
used in older children and adults because their sera may tration of proteins and is affected very little by electrolytes
Liver Function Tests 59

and other molecules. Protein concentration is read 1. In cirrhosis, albumin may be reduced and there may
directly from the scale on the refractometer. In biuret be polyclonal increase of IgG and IgA, with β-γ
method, copper ions react with peptide bonds of proteins bridging. (IgA migrates between β and γ regions which
and form a violet-colored compound. Intensity of this obscures the demarcation between β and γ peaks).
color (measured colorimetrically) is proportional to the 2. In primary biliary cirrhosis, there is polyclonal
concentration of proteins. increase of IgM.
Total serum protein level is affected by both albumin 3. In α1-antitrypsin deficiency (associated with cirrhosis)
and gamma globulins. In cirrhosis, decrease in albumin α1- globulin band is reduced.
level is often compensated by increase in the level of 4. In chronic active hepatitis, IgG is elevated.
gamma globulins; therefore, estimation of total serum
proteins is of limited value in cirrhosis. Estimation of Prothrombin Time (PT)
serum albumin and serum protein electrophoresis are Most of the coagulation proteins are synthesized in the
more helpful. liver. Vitamin K is required for the synthesis of factors
II, VII, IX, and X by the hepatocytes; therefore these
2. Serum albumin: Albumin is synthesized exclusively
factors are called as vitamin K-dependent factors.
in liver and constitutes about 60% of total proteins in
Synthesis of these factors is deficient in hepatocellular
serum; therefore its estimation is an important investi-
disease. In obstructive jaundice, vitamin K (a fat-soluble
gation in liver disease. Half-life of albumin is about 20
vitamin) cannot be absorbed due to the absence of bile
days and therefore fall in its level in response to decreased
in the intestine.
synthesis is not immediately apparent. Therefore, in
PT measures three out of four vitamin K-dependent
acute liver disease (e.g. viral hepatitis), there is little
factors (II, VII, and X) and is prolonged in hepatocellular
change in albumin level. Serum albumin level is low in
disease and in obstructive jaundice. Intramuscular
chronic liver disease (cirrhosis) and correlates with
injection of vitamin K corrects prolonged PT in
synthetic capacity of hepatocytes; therefore, it is helpful
obstructive jaundice but not in hepatocellular jaundice.
in following progression of cirrhosis. Also, fall in serum
In acute fulminant liver failure, marked prolongation
albumin level correlates with severity of ascites. In
of PT is an unfavourable prognostic sign.
cirrhosis and in chronic active hepatitis, serum gamma
globulins are increased due to inflammation. Low To distinguish between a prolonged PT due to hepato-
albumin and raised gamma globulins in serum cause cellular disease from that due to cholestasis with fat
reversal of albumin/globulin ratio. malabsorption, PT is repeated after administration of
Serum albumin is estimated by bromocresol green vitamin K. Reduction of prolonged PT occurs in
method. Bromocresol green is an indicator dye, which cholestatic liver disease, but not in hepatocellular
when added to serum, binds selectively and tightly to disease.
albumin and becomes blue in color. Absorbance (in a
spectrophotometer at 632 nm) is directly proportional to Blood Ammonia
concentration of albumin. Blood ammonia is mainly derived from gastrointestinal
Causes of decreased serum albumin: tract. In the intestine, bacterial enzymes act on nitrogen-
• Decreased intake: malnutrition. containing foods to produce ammonia, which is carried
• Decreased absorption: malabsorption syndromes. to the liver via portal vein. In the liver, ammonia is
• Decreased synthesis: liver disease, chronic infections. converted to non-toxic urea in the urea cycle.
• Increased catabolism: thyrotoxicosis, fever, malig- Increased blood ammonia levels are seen in:
nancy, infections. • Fulminant hepatic failure
• Increased loss: nephrotic syndrome, severe burns, • Cirrhosis
protein-losing enteropathies, ascites • Reye’s syndrome
• Increased blood volume: pregnancy, congestive • “Shunting” of portal blood to systemic circulation
cardiac failure. • Gastrointestinal hemorrhage (there is increased
As low serum albumin occurs in diseases other than production of ammonia from blood proteins by
those of liver, serum albumin is a sensitive but non- bacterial enzymes). In hepatic disease, gastro-
specific test for liver disease. intestinal hemorrhage is associated with increased
3. Serum protein electrophoresis: Details of serum risk of hepatic encephalopathy.
protein electrophoresis are given in Chapter 28 • Inherited deficiencies of urea cycle enzymes.
“Laboratory Tests in Hematological Malignancies”. If raised, estimation of blood ammonia is likely to be
In liver disease, following changes may be seen on helpful in patients with coma of unknown origin, since
protein electrophoresis (Fig. 4.6): it is indicative of hepatic encephalopathy.
60 Essentials of Clinical Pathology

Fig. 4.6: Serum protein electrophoresis patterns and densitometer scans in normal individuals and in
cirrhosis of liver and α1-antitrypsin deficiency

Tests which Assess Hepatic Injury • 5’-nucleotidase (5’-NT)


(Liver Enzyme Studies) Locations of enzymes in liver cell are shown in Figure
Serum enzyme changes in liver disease result from 4.7. Degree of elevation of a particular enzyme depends
hepatocyte damage and do not indicate hepatic on pattern of hepatic damage.
functional capacity. In the investigation of liver disease,
following serum enzymes are measured: Serum Aminotransferases
• Serum aspartate aminotransferase or AST (formerly
called serum glutamic-oxaloacetic transaminase or Serum aminotransferases are the sensitive markers of
SGOT) acute hepatocellular injury. ALT is a cytosolic enzyme
• Serum alanine aminotransferase or ALT (formerly while AST is both cytosolic and mitochondrial.
called serum glutamic-pyruvic transaminase or Normally, aminotransferases are present in serum at
SGPT) a low level. When necrosis or death of cells containing
• Serum alkaline phosphatase or ALP these enzymes occurs, aminotransferases are released
• γ-Glutamyl transferase or GGT (also called as into the blood and their concentration in blood increases.
γ-glutamyl transpeptidase) This level correlates with extent of tissue damage.
Liver Function Tests 61

Most marked elevations of ALT and AST (>15 times Serum Alkaline Phosphatase (ALP)
normal) are seen in acute viral hepatitis, toxin-induced
Alkaline phosphatase is distributed widely in various
hepatocellular damage (e.g. carbon tetrachloride), and
tissues like liver, bones, intestine, kidney, and placenta.
centrilobular necrosis due to ischemia (congestive
In the liver, ALP, GGT, and 5’-NT are located normally
cardiac failure).
on canalicular surface of hepatocytes (See Fig. 4.7). In
Moderate elevations (5-15 times) occur in chronic
cholestasis, accumulated bile acids dissolve canalicular
hepatitis, autoimmune hepatitis, alcoholic hepatitis, acute side of hepatocyte membrane and enzymes are released
biliary tract obstruction, and drug-induced hepatitis. in blood. Therefore, diseases that affect mainly
Mild elevations (1-3 times) are seen in cirrhosis, non- hepatocyte secretion have elevated levels of ALP.
alcoholic steatosis, and cholestasis. Measurement of ALP is helpful in differentiation of
Determinations of these enzymes are helpful in the hepatocellular jaundice from cholestatic jaundice
differential diagnosis of hepatocellular from cholestatic (See Fig. 4.8).
jaundice. Increase of AST and ALT is much more in
Causes of increased ALP
hepatocellular jaundice (>500 units/ml) than in choles-
1. Hepatobiliary disease: ALP is increased in most cases
tatic jaundice (<200 units/ml) (Fig. 4.8).
of cholestatic type of jaundice. While hepatocellular
Although determination of any one aminotransferase
injury is characterized by marked elevation of ALT
is adequate, measurement of both can be helpful in the and AST, cholestasis is characterized by marked
calculation of AST/ALT ratio. Normal ratio is 0.7 to 1.4. increase (more than 3 times normal) of ALP. Since
Increased ratio (>2.0) is highly suggestive of alcoholic there are many other sources of ALP apart from liver,
hepatitis, while ratio <1.0 is seen in acute viral hepatitis. simultaneous measurement of serum GGT and
ALT and AST are elevated in acute viral hepatitis serum 5’-NT may be used to ascertain whether
even before the appearance of jaundice. Persistence of increase of ALP is of hepatic origin.
elevated ALT and AST beyond 6 months in a case of Hepatobiliary causes of increased alkaline
hepatitis indicates development of chronic hepatitis. phosphatase are:
In massive liver necrosis, aminotransferase levels • Bile duct obstruction (cancer of head of pancreas,
gradually decrease. Therefore, falling levels do not stone in common bile duct, stricture of bile duct,
necessarily indicate recovery from acute hepatitis. biliary atresia)

Fig. 4.7: Locations of enzymes in liver cell. The enzymes are found in specific locations as follows: Alanine aminotransferase
(ALT): cytosol; Aspartate aminotransferase (AST): mitochondria and cytosol; Lactate dehydrogenase (LDH): cytosol; Alkaline
phosphatase (ALP): canalicular surface; Gamma glutamyl transferase (GGT): canalicular surface and microsomes; 5’ NT:
canalicular surface. The pattern of hepatocyte injury determines the enzymes elevated: cytoplasmic damage: elevated AST,
ALT, and LDH; mitochondrial damage: elevated AST; cholestatic damage: elevated ALP and GGT
62 Essentials of Clinical Pathology

Fig. 4.8: (A) Comparative serum alanine transferase (ALT) levels in liver diseases. Red horizontal line represents upper limit
of normal. (B) Comparison of percentage levels of serum alkaline phosphatase (ALP) in hepatocellular disease and cholestasis.
Red horizontal line represents discriminatory level between hepatocellular disease and cholestasis (i.e. 3 times normal), while
black horizontal line represents upper limit of normal

• Primary biliary cirrhosis Serum γ-glutamyl Transferase (GGT)


• Primary sclerosing cholangitis This is also called as γ-glutamyl transpeptidase. Relatively
• Infiltrative diseases of liver (granulomatous
high levels of this enzyme are present in liver, pancreas,
diseases like tuberculosis or sarcoidosis, amyloi-
kidney, and prostate. Estimation of this enzyme is
dosis, cysts, primary or secondary cancer)
Further work-up is necessary to diagnose the particularly useful in following liver diseases:
cause of cholestasis depending on history and 1. Alcoholism: Increased enzyme activity is present in
physical examination findings. This usually consists alcoholism, and is a helpful clue in suspected cases
of abdominal ultrasonography or CT scan (for of occult alcoholism (even in the absence of alcoholic
detection of dilated bile ducts which indicate liver disease). It is also helpful in follow up of patients
extrahepatic obstruction, or for detection of focal or with chronic alcoholism. Marked elevation of GGT
diffuse parenchymal lesion of liver); endoscopic occurs in acute alcoholic hepatitis.
retrograde cholangiopancreatography (ERCP) or 2. Cholestasis: Elevation of GGT generally parallels that
percutaneous transhepatic cholangiography (PTC) to of ALP and 5’-NT in liver disease. Elevation of ALP
diagnose cause of biliary obstruction; and liver is not specific for liver disease. Elevation of both ALP
biopsy. and GGT points towards liver disease.
2. Diseases of bones: ALP is present within osteoblasts.
3. Recovery in acute hepatitis: Serum GGT is the last
Due to high osteoblastic activity during active bone
enzyme to return to normal following acute hepatitis
growth, serum ALP is higher in children than in
and its normalization is indicative of a favourable
adults. Elevation of ALP occurs in conditions with
increased osteoblastic activity like osteomalacia, outcome.
rickets, hyperparathyroidism, Paget’s disease,
5’-nucleotidase (5’-NT)
osteosarcoma, and osteoblastic type of metastatic
carcinoma. 5’-NT is present in liver as well as in various other tissues.
3. Pregnancy: Serum ALP is increased during It is located mainly along the cell membrane, similar to
pregnancy due to secretion from placenta. ALP and GGT. Estimation of 5’-NT is helpful in deciding

tahir99 - UnitedVRG
vip.persianss.ir
Liver Function Tests 63

whether increased ALP is due to liver disease or due to impairment of bile flow that may be intrahepatic or
increased osteoblastic activity in growing children. extrahepatic). History and physical examination findings
should be correlated with liver function tests.
Tests that Assess Clearance of Exogenous
Substances from the Liver Box 4.4: Typical LFT profile in hepatocellular disease
Some synthetic dyes, when introduced into the body, are • Marked elevation of AST and ALT (usually >500 IU)
rapidly removed from the blood stream by the liver and • Mild increase of ALP (<3 times normal)
excreted into the bile. Disappearance of these dyes from • Hyperbilirubinemia, if present, is of both conjugated and
the blood is dependent on adequate hepatic circulation, unconjugated type
normal hepatocyte function, and uninterrupted bile flow.
Dye excretion tests are sensitive tests of liver function Hepatocellular injury may be acute (e.g. acute viral
and are usually indicated in those patients in whom liver hepatitis, toxic hepatitis, ischemic hepatitis, alcoholic
disease is suspected but have little or no jaundice. Three hepatitis) or chronic (cirrhosis, chronic active hepatitis,
synthetic dyes are commonly employed to test liver autoimmune hepatitis). Acute hepatocellular injury is
function: bromosulphthalein (BSP), indocyanine green usually associated with marked elevation of ALT and
(used by surgeons before resecting liver for hepato- AST and mild elevation of alkaline phosphatase (Box 4.4).
cellular cancer) , and Rose Bengal. However, because of In chronic hepatocellular injury, aminotransferases are
the risk of adverse reactions and advent of more sensitive moderately elevated, and serum albumin is reduced.
tests for detection of liver disease, dye excretion tests are Cholestasis is characterized by mild elevation of ALT
nowadays rarely used. and AST and marked increase of ALP and GGT (Box
Bromosulphthalein (BSP) excretion test: After adminis- 4.5). With complete obstruction of bile ducts, both serum
tration, BSP is taken up by the hepatocytes, binds to ALP and serum bilirubin are increased, stools are pale,
ligandin and Z protein, conjugated to glutathione, and and urine contains no urobilinogen. The pattern of
then excreted into the bile. In this test, BSP is injected elevated ALP but normal serum bilirubin is seen in
intravenously, a blood specimen is obtained after a infiltrative diseases. Laboratory abnormalities often
specified time, and the percent of dye retained in blood overlap in cholestatic and infiltrative diseases; imaging
is calculated. If the amount of dye retained in blood is studies (such as ultrasound, CT scan, magnetic resonance
more than 50% at 45 minutes, abnormal liver function is imaging, and cholangiography) and liver biopsy are
present. needed for distinction between them.
BSP excretion test will yield falsely abnormal result
if hepatic circulation is impaired. Also, there is a risk of Box 4.5: Typical LFT profile in cholestatic jaundice
adverse reactions including tissue necrosis and anaphy- • Marked elevation of ALP (>3 times normal)
laxis in a small number of patients. • Elevation of GGT and 5’-NT
BSP test remains useful in the diagnosis of Dubin- • Mild or no increase of ALT and AST (usually <200 IU)
Johnson syndrome and its differentiation from Rotor • Elevation of conjugated bilirubin
syndrome. A higher level of BSP in blood at 2 hours with
normal value at 45 minutes is seen in Dubin-Johnson For specific or etiologic diagnosis of liver disease,
syndrome. In Rotor syndrome, value of BSP is higher at further testing is required:
45 minutes and lower at 2 hours. • Hepatocellular disease: In patients with hepato-
cellular pattern of injury, common investigations to
INTERPRETATION OF detect underlying cause include viral serology (viral
LIVER FUNCTION TESTS hepatitis: IgM anti-hepatitis A antibody, hepatitis
A stepwise approach consists of laboratory tests for: surface B antigen (HBsAg), antihepatitis C antibody),
• Presence of liver disease: History, physical findings, search for injurious drugs, toxins or alcohol,
and liver function tests autoantibodies like antinuclear antibodies and anti-
• Nature of liver disease: whether hepatocellular (cell smooth muscle antibodies (autoimmune hepatitis),
injury) or cholestatic and serum ceruloplasmin (Wilson disease). If cause
• Specific cause of liver disease is not detected in the presence of persistent elevation
• Assessment of severity of liver disease of aminotransferases, liver biopsy is performed (that
Liver diseases can be broadly classified into two types: may reveal chronic viral hepatitis, autoimmune
hepatocellular (primary abnormality is liver cell disorders, Wilson disease, haemochromatosis, and
necrosis), and cholestatic (primary abnormality is infiltrative diseases) (Table 4.4 and Figure 4.9).

tahir99 - UnitedVRG
vip.persianss.ir
64 Essentials of Clinical Pathology

Table 4.4: Differentiation of causes of raised aminotransferases


Cause Clinical features Diagnosis

1. Viral hepatitis A H/o exposure in endemic areas IgM anti-hepatitis A antibodies


2. Viral hepatitis B H/o nonsterile injections, IV drug IgM anti-hepatitis B core antigen
abuse, blood transfusion, multiple antibodies, hepatitis B surface antigen
sexual partners, homosexuals
3. Viral hepatitis C As for hepatitis B Hepatitis C-RNA, antihepatitis C
antibodies
4. Viral hepatitis E H/o exposure in endemic area Anti-HEV
5. Alcoholic liver disease H/o alcohol abuse AST/ALT ratio > 2.0, ↑ GGT
6. Nonalcoholic steatohepatitis Type 2 DM, obesity, hyperlipidemia Liver biopsy
7. Autoimmune hepatitis Young female Raised immunoglobulins, low albumin,
antinuclear antibody+, anti-smooth
muscle antibody+, liver kidney
microsomal antibody+
8. Hemochromatosis Autosomal recessive, diabetes mellitus, Transferrin saturation >45%, genetic
skin pigmentation, chronic liver disease, testing
multiorgan dysfunction
9. Wilson’s disease Young age, autosomal recessive, Low ALP, high serum bilirubin, low serum
Kayser-Fleischer ring, hemolytic ceruloplasmin
anemia

Fig. 4.9: Evaluation of acute hepatocellular injury

• In patients with cholestatic or infiltrative pattern of LIVER BIOPSY


injury, imaging studies should be done for diagnosis
Liver biopsy is a procedure in which a small piece of
of obstruction. If there is no evidence of obstruction,
liver tissue is removed and examined microscopically to
liver biopsy is usually done (Table 4.5 and Fig. 4.10).
Severity of liver disease is assessed by serum determine the cause and severity of liver disease. Paul
bilirubin, serum albumin, and prothrombin time. Ehrlich performed the first percutaneous liver biopsy in
Child-Turcotte-Pugh classification is commonly used to 1883 in Germany. Since then the technique has been
assess severity of cirrhosis and is based on both clinical modified and now a variety of approaches can be used
and laboratory parameters (Table 4.6). to obtain a liver biopsy sample. Before proceeding with

tahir99 - UnitedVRG
vip.persianss.ir
Liver Function Tests 65

Table 4.5: Differentiation of causes of raised serum alkaline phosphatase in jaundice

Cause Clinical features Investigations

1. Primary biliary cirrhosis Middle-aged female; pruritus; Antimitochondrial antibody; raised


xanthelasma serum cholesterol
2. Primary sclerosing cholangitis Adult male; associated inflammatory Antinuclear cytoplasmic antibody,
bowel disease endoscopic retrograde
cholangiopancreatography
3. Extrahepatic biliary obstruction Dark urine; clay-colored stools; Ultrasonography for dilated bile ducts;
palpable gallbladder ± endoscopic retrograde
cholangiopancreatography

Fig. 4.10: Evaluation of raised serum alkaline phosphatase in cholestatic injury

the biopsy, potential risks of the procedure and benefits TYPES OF LIVER BIOPSY
of histological examination should be assessed and the Currently, several methods are available for obtaining
procedure should be performed only when benefits out liver tissue specimen. Choice of the method depends on
weigh the risks. its availability, clinical situation, and preference of the

tahir99 - UnitedVRG
vip.persianss.ir
66 Essentials of Clinical Pathology

Table 4.6: Child-Turcotte-Pugh grading system to assess severity of cirrhosis

Parameter Score 1 Score 2 Score 3

Serum bilirubin (mg/dl) <2 2-3 >3


Serum albumin (g/dl) ≥ 3.5 2.8-3.5 <2.8
Prothrombin time (difference in seconds) <4 4-6 >6
Ascites None Slight or controlled by diuretics Moderate despite diuretics
Encephalopathy (grade) 0 1-2 3-4

Grade A (excellent prognosis): Score 5 or 6; Grade B (intermediate prognosis): Score 7 to 9; Grade C (very poor prognosis):
Score ≥ 10.

operator. An experienced clinician, gastroenterologist, 2. Chronic liver disease:


or hepatologist performs the liver biopsy. A radiologist • To define cause
performs transjugular biopsy. • Grading of inflammatory activity
Methods of obtaining liver biopsy are: • Staging (prognosis) of chronic hepatitis C and
• Percutaneous liver biopsy chronic hepatitis B
– Percutaneous ‘blind’ liver biopsy • Monitor response to therapy
– Percutaneous guided liver biopsy 3. Diagnosis of metabolic liver disorders like
– Percutaneous plugged liver biopsy haemochromatosis and Wilson’s disease and
• Transvenous (Transjugular) liver biopsy quantitation of accumulated iron and copper
• Laparoscopic liver biopsy levels respectively.
4. Diagnosis and grading of alcoholic liver disease
Percutaneous ‘Blind’ Liver Biopsy 5. Accurate staging of advanced cases of primary
In this technique, liver biopsy is carried out without any biliary cirrhosis and primary sclerosing cholan-
real time imaging guidance. Borders of the liver are gitis.
defined by percussion and the biopsy needle is inserted 6. Investigation of persistently abnormal and
either through the intercostal or subcostal route. unexplained LFTs.
Subcostal route is employed if liver is enlarged below 7. Investigation of pyrexia of unknown origin.
the costal margin. 8. Diagnosis of infections like tuberculosis.
Liver biopsy needles are of three main types: suction 9. Diagnosis of focal hepatic lesions
needles (Menghini, Jamshidi), cutting needles (Tru-Cut, 10. Histologic monitoring following liver trans-
Vim-Silverman) (Fig. 4.11), and spring-loaded cutting plantation.
needles. Cutting needles require longer time for biopsy
Contraindications:
as compared to the suction needles and remain in liver
• Uncooperative patient: If patient is not cooperative,
for a longer period, thus increasing the risk of bleeding.
movement during the procedure can lead to lacera-
Suction needles often cause fragmentation of cirrhotic
tion of liver and bleeding.
liver tissue.
• Extrahepatic biliary obstruction: Injury to dilated bile
Indications: ducts can lead to biliary peritonitis.
1. Acute hepatitis of unknown cause: (Liver biopsy • Bleeding diathesis or abnormal coagulation profile:
is not required in typical acute viral hepatitis). Prolongation of prothrombin time >3-5 seconds above
control value, platelet count < 50,000/ml, or bleeding
time ≥10 minutes are considered as contraindications
to liver biopsy. If liver biopsy is essential, then biopsy
can be performed after administration of vitamin K,
fresh frozen plasma, or platelet concentrate. Alter-
natively, transjugular approach can be used.
In patients with hemophilia who are infected
with hepatitis C or B virus through blood transfusion,
liver biopsy can be carried out to assess liver damage
Fig. 4.11: Vim-Silverman liver biopsy needle after correcting coagulation deficiency.

tahir99 - UnitedVRG
vip.persianss.ir
Liver Function Tests 67

• Tense ascites: Tense ascites is associated with failure real time imaging of the liver (ultrasonography,
to obtain liver tissue (due to increased distance computed tomography, or magnetic resonance imaging).
between abdominal wall and liver) and risk of This approach is suitable for biopsy of focal lesions.
bleeding in ascites. Biopsy can be performed after
removal of ascitic fluid or through transjugular Percutaneous ‘Plugged’ Liver Biopsy
approach.
In this method, after taking the biopsy sample, the outer
• Hydatid cyst of liver: Puncture of hydatid cyst will
sheath of the needle is left in place and the obturator
lead to spread of cysts throughout the abdomen and
holding the liver biopsy tissue is removed. A cannula is
sometimes, anaphylactic reaction.
then introduced through the outer sheath and gel or gel
• Suspected hemangioma or other highly vascular
foam is injected to seal or ‘plug’ the needle track in the
tumor
liver. This method is said to reduce the risk of bleeding
• Amyloidosis is associated with increased risk of
in individuals with impaired coagulation.
bleeding.
Patients with encephalopathy, hepatic failure with
Transvenous (Transjugular) Liver Biopsy
severe jaundice, severe congestive cardiac failure, and
advanced age are at increased risk of complications Liver biopsy is obtained from within the vascular system
following liver biopsy. of liver. A small catheter is inserted through the jugular
vein in the neck and radiologically guided, via right
Pre-requisites:
atrium and inferior vena cava, into the hepatic vein. The
• Prior imaging of liver should be carried out to identify
biopsy needle is then inserted through the catheter,
any abnormality, to define borders, and to determine
advanced into the liver, and biopsy is taken.
relative positions of gallbladder, lungs, and kidneys.
This method is suitable in severe coagulation
• Routine hemogram
disorders and in massive ascites. Due to the risk of cardiac
• Coagulation profile
arrhythmias (as the catheter passes through the atrium),
• Informed consent
close cardiac monitoring is required during this
• Blood grouping and cross matching
procedure.
Method:
1. Patient lies in supine position. Laparoscopic Liver Biopsy
2. After selecting the site for liver biopsy, a local
A laparoscope is introduced through an incision in the
anesthetic is injected.
abdominal wall, and liver biopsy is obtained under direct
3. A small incision is made and the biopsy needle is
visualization. Usually, such a biopsy is taken when a focal
passed into the liver. Patient is asked to hold his/her
lesion is incidentally detected on diagnostic laparoscopy
breath in expiration. Method of obtaining liver tissue
of abdomen.
depends on the type of needle used.
4. After needle is removed, patient lies supine or on the
right side and is closely monitored especially during
REFERENCE RANGES
first 6 hours for early detection of complications. Serum alanine aminotransferase (ALT, SGPT): 5-42 U/L
Complications: Serum aspartate aminotransferase (AST, SGOT): 5-40 U/L
1. Pain Serum alkaline phosphatase (ALP):
2. Intraperitoneal hemorrhage: Patients at particular • Children: 25-350 U/L
risk of bleeding are those with cirrhosis and malig- • Adult males: 25-120 U/L
nancy in liver. These patients should not be biopsied • Adult females: 25-90 U/L
on outpatient basis. AST/ALT ratio: 0.7-1.4
3. Biliary peritonitis due to puncture of gallbladder. Serum bilirubin:
4. Puncture of other organs like kidney, lung, and colon. • Total: 0.3-1.0 mg/dl
Overall mortality following liver biopsy is reported • Direct (Conjugated): 0-0.2 mg/dl
to be 0.1%. Serum proteins, total: 5.5-8.0 gm/dl
Serum albumin: 3.5-5.0 gm/dl
Percutaneous Guided Liver Biopsy
Serum globulins: 1.8-3.5 gm/dl
In percutaneous guided liver biopsy, needle is inserted
into the liver through the abdomen or lower chest during Albumin/Globulin (A/G) ratio: >1.5

tahir99 - UnitedVRG
vip.persianss.ir
68 Essentials of Clinical Pathology

Serum protein electrophoresis: 6. Dufour DR, Lott JA, Nolte FS, Gretch DR, Koff RS, Seeff
Albumin 52-65% LB. Diagnosis and monitoring of hepatic injury. II.
α1 globulin: 2.5-5% Recommendations for use of laboratory tests in
α2 globulin: 7-13% screening, diagnosis, and monitoring. Clin Chem 2000;
β globulin: 8-14% 46:2050-68.
γ globulin: 12-22% 7. Dufour DR, Lott JA, Nolte FS, Gretch DR, Koff RS, Seeff
LB. Diagnosis and monitoring of hepatic injury. I.
Prothrombin time: 11-15 seconds Performance characteristics of laboratory tests. Clin
Chem 2000;46:2027-49.
Plasma ammonia: 9-33 μmol/L
8. Gaw A, Murphy MJ, Cowan RA, O’Reilly DSJ, Stewart
Serum gammaglutamyl transferase: MJ, Shepherd J. Clinical Biochemistry. An Illustrated
• Males: Up to 40 U/L Colour Text, 3rd Ed. Edinburgh. Churchill Livingstone.
• Females: Up to 25 U/L 2004.
9. Grant A, Neuberger J. Guidelines on the use of liver
CRITICAL VALUES biopsy in clinical practice. Gut 1999; 45 (Suppl IV):
IV1-IV11.
Plasma ammonia: >40 μmol/L 10. Johnston DE. Special considerations in interpreting liver
function tests. Am Fam Physician 1999;59:2223-30.
Serum bilirubin: >15 mg/dl in newborns 11. Limdi JK, Hyde GM. Evaluation of abnormal liver
function tests. Postgrad Med J 2003;79;307-12.
BIBLIOGRAPHY 12. Lucey MR, Brown KA, Everson GT, et al. Minimal
criteria for placement of adults on the liver transplant
1. American Gastroenterological Association Clinical
waiting list; a report of a national conference organized
Practice Committee: AGA technical review on liver
by the American Society of Transplant Physicians and
chemistry tests. Gastroenterology 2002;123:1367-84.
2. American Gastroenterological Association Medical the American Association for the Study of Liver
Position Statement: Evaluation of liver chemistry tests. Disease. Liver Transpl Surg 1997;3:628-37.
Gastroenterology 2002;123:1364-6 13. Pugh RN, Murray-Lyon IM, Dawson JL, et al.
3. Beckingham IJ, Ryder SD. Investigation of liver and Transection of the oesophagus for bleeding oesopha-
biliary disease. BMJ 2001;322:33-6. geal varices. Br J Surg 1973;60:646-9.
4. Black ER. Diagnostic strategies and test algorithms in 14. Roche SP, Kobos R. Jaundice in the adult patient. Am
liver disease. Clin Chem 1997;43:1555-60. Fam Physician 2003;69:299-304.
5. Burke MD. Liver function: test selection and interpreta- 15. Thapa BR, Walia A. Liver function tests and their
tion of results. Clin Lab Med 2002;22:377-90. interpretation. Indian J Pediatr 2007;74:663-71.

tahir99 - UnitedVRG
vip.persianss.ir
5

Disorders of Lipids and


Biochemical Cardiac Markers

DISORDERS OF LIPIDS triglycerides, fat-soluble vitamins) that is surrounded by


(ii) a surface monolayer composed of phospholipids, free
The major lipids present in blood are cholesterol, fatty
cholesterol, and apoproteins (Fig. 5.1). The surface of
acids, and triglycerides. Lipid disorders are common in
clinical practice, and some of them are associated with lipoproteins is water-soluble or polar while core is
an increased risk of atherosclerotic cardiovascular hydrophobic or non-polar.
disease. Cardiovascular disease is a major cause of There are five major lipoprotein classes: chylo-
mortality in persons under the age of 60, and proper microns, very low-density lipoproteins (VLDL), inter-
management of lipid abnormalities significantly reduces mediate density lipoproteins (IDL), low-density lipo-
this risk. proteins (LDL), and high-density lipoproteins (HDL)
Lipids are insoluble in plasma and are therefore (Fig. 5.2 and Table 5.1). Triglycerides are carried mainly
transported in circulation in association with proteins. by chylomicrons, VLDL, and LDL, while major lipo-
These complexes of lipids and proteins are known as proteins for cholesterol transport are LDL and HDL.
lipoproteins. Dyslipidemias are disorders of lipoprotein
metabolism. Chylomicrons: These are the largest and the least dense
of the lipoproteins, and transport exogenous lipids to
PHYSIOLOGY various cells. Dietary fat is incorporated into chylo-
microns by intestinal epithelial cells. The lipid core of
Cholesterol and Triglycerides
The two major lipids in blood are cholesterol and
triglycerides. Since they are insoluble in water, they are
carried by lipoproteins.
Cholesterol is a lipid found in all cell membranes and
in blood plasma. Cholesterol is an essential component
of the cell membranes, and is necessary for synthesis of
steroid hormones, and for the formation of bile acids.
Cholesterol is synthesized by liver and many other
organs, and is also ingested in the diet.
Triglycerides are lipids in which three long-chain fatty
acids are attached to glycerol. Triglycerides serve as a
source of energy. They are present in dietary fat and also Fig. 5.1: Basic structure of a lipoprotein molecule. Lipoproteins
synthesized by liver and adipose tissue. are spherical aggregates of lipids and apolipoproteins. They
consist of a core of triglycerides and cholesterol esters
Lipoproteins surrounded by a shell of phospholipids and cholesterol.
Apolipoproteins are embedded in the shell. The larger the lipid
Cholesterol and triglycerides are not soluble in water and core, the lower is its density. Lipoproteins are classified into
are transported in blood incorporated into lipoproteins. 5 types: chylomicrons, very low density lipoprotein (VLDL),
Lipoproteins are spherical macromolecular complexes intermediate density lipoprotein (IDL), low density lipoprotein
consisting of (i) a central core of lipids (cholesterol ester, (LDL), and high density lipoprotein (HDL)

tahir99 - UnitedVRG
vip.persianss.ir
70 Essentials of Clinical Pathology

Fig. 5.2: Lipoproteins and their composition

Table 5.1: Summary of major lipoproteins

Lipoprotein Major lipid Formation Function


carried

1. Chylomicron Triglyceride Secreted by intestinal Transport of exogenous triglycerides, cholesterol,


epithelial cells and fat-soluble vitamins absorbed from food to
the peripheral tissues

2. Very low density Triglycerides, Secreted by liver Transport of endogenous triglycerides to adipose
lipoprotein Cholesterol tissue and muscle from liver
3. Low density Cholesterol Formed from modification Transport of cholesterol from liver to peripheral
lipoprotein of VLDL by lipoprotein tissues
lipase in peripheral
tissues
4. High density Cholesterol Secreted from liver “Reverse cholesterol transport”, i.e. from peripheral
lipoprotein tissues to liver

chylomicrons consists mainly of triglycerides, some cholesterol. It carries most of the endogenous
cholesterol and fat-soluble vitamins. Intestinal cells triglyceride from liver to adipose tissue and muscle.
secrete chylomicrons into the lymphatics, which then Triglyceride is removed by the action of lipoprotein lipase
enter the bloodstream via the thoracic duct. In circulation, in the circulation and VLDL particle becomes smaller,
chylomicrons are acted upon by lipoprotein lipase to when it is called as intermediate density lipoprotein
release triglycerides; further hydrolysis of triglycerides (IDL). Further processing of IDL leads to the formation
by lipoprotein lipase causes release of free fatty acids that of low-density lipoprotein (LDL), which is the major
are then taken up by adipose tissue and muscle. Liver
carrier for cholesterol.
takes up the cholesterol-rich chylomicron remnant
particle and cholesterol enters the metabolic pathway. Intermediate density lipoprotein (IDL): This is the
Very low-density lipoproteins (VLDL): VLDL particle remnant of VLDL formed when triglycerides are
is synthesized by the liver. It transports triglycerides and removed from VLDL by lipoprotein lipase in circulation.

tahir99 - UnitedVRG
vip.persianss.ir
Disorders of Lipids and Biochemical Cardiac Markers 71

About half of IDL is cleared from blood by the liver and cholesterol esters and incorporate them into the core of
the remaining half is further processed to form LDL. the chylomicrons. Chylomicrons are secreted into the
Normally, IDL is not detected in plasma as it is formed lymphatics from where they reach the bloodstream via
transiently. thoracic duct. In the circulation, endothelial-bound
lipoprotein lipase hydrolyzes triglycerides in the
Low-density lipoprotein (LDL): LDL is the major carrier chylomicron core and releases free fatty acids that are
lipoprotein for cholesterol from liver to the peripheral then taken up by the adipose tissue and muscle; in these
tissues. It is formed from VLDL. LDL plays a major role the free fatty acids are converted to triglycerides and
in the genesis of atherosclerosis. stored. The remaining smaller chylomicron is taken by
LDL is taken up by the cells through the LDL receptor, the liver, degraded, and cholesterol contained therein is
a glycoprotein. The LDL receptor is present on the surface then used for the formation of bile acids, incorporated
of all cells and recognizes apolipoprotein B on the surface into cell membranes, secreted in blood as lipoprotein
of LDL particle. After internalization of LDL particle, the cholesterol, or excreted in bile (Fig. 5.3).
lipoprotein is catabolized and the receptor is recycled
back to the cell surface. Intracellularly, LDL is degraded Endogenous pathway: This pathway is divided into:
to free cholesterol that is needed for cellular needs. The • Apo B-100 lipoprotein system
level of LDL in circulation is determined by number and • Apo A-I lipoprotein system
function of LDL receptors. Joseph Goldstein and Michael Apo B-100 lipoprotein system: In the liver, triglycerides and
Brown were awarded the Nobel Prize for Physiology or cholesterol are assembled with apo B-100 and
Medicine in 1985 for characterizing the LDL receptor. phospholipids to produce VLDL. VLDL represents the
Genetic absence of LDL receptors leads to familial hyper- major export pathway for cholesterol from liver. After
cholesterolemia. its secretion from the liver, lipoprotein lipase on capillary
High-density lipoprotein (HDL): HDL binds to endothelium hydrolyzes triglyceride in the core of the
peripheral tissues that have apolipoprotein A receptors VLDL particles resulting in the formation of (cholesterol
and takes up cholesterol. HDL cholesterol is either taken ester-rich) intermediate density lipoproteins (IDL).
by the liver or is incorporated into IDL to form LDL. Further degradation results in the formation of LDL
particles that are rich in cholesterol ester. LDL particles
Lipoprotein (a) or Lp (a): Attachment of apolipoprotein
are taken up by all nucleated cells through LDL receptors
(a) molecule to apolipoprotein B molecule on the surface
(receptor- mediated endocytosis) or by other scavenger
of LDL particle leads to the formation of a new particle
routes (e.g. monocytes or foam cell in atheromatous
called as lipoprotein (a). Excess Lp (a) is associated with plaques).
risk of atherosclerosis.
Apo A-I lipoprotein system: High-density lipoprotein
Lipoprotein Metabolism (HDL) particles are synthesized by liver and intestine
and participate in reverse cholesterol transport. HDL, rich
There are two pathways of lipoprotein metabolism:
in apo A-I, acquires free cholesterol from peripheral
exogenous and endogenous.
tissues, esterifies it, and either transfers it directly to the
Exogenous pathway: Small intestinal cells absorb fatty liver or to other lipoproteins (IDL and LDL), which then
acids and cholesterol, esterify them into triglycerides and transport it to liver (Fig. 5.4).

Fig. 5.3: Exogenous lipid pathway showing route of metabolism of lipid absorbed from the intestine

tahir99 - UnitedVRG
vip.persianss.ir
72 Essentials of Clinical Pathology

Fig. 5.4: Endogenous lipid pathway showing route of metabolism of endogenously synthesized lipid Abbreviations: VLDL:
very low density lipoprotein; IDL: intermediate density lipoprotein; LDL: Low density lipoprotein; HDL: high density lipoprotein;
LCAT: lecithin-cholesterol acyltransferase

Apolipoproteins commonly classified according to the Fredrickson or


World Health Organization classification (Table 5.2). This
These are proteins located on the surface of lipoproteins.
classification is based on the particular pattern of lipid
They aid in lipid transport and delivery in three ways:
and lipoprotein abnormality. On electrophorsis, four
(i) stabilizing structure of lipoproteins, (ii) serving as
principal bands are observed from cathode (–) to anode
regulatory cofactors for enzymes that act on lipoproteins,
(+): chylomicrons, β (LDL), pre β (VLDL), and α (HDL)
and (iii) acting as ligands for binding to lipoprotein
(Fig. 5.5). Fredrickson classification of hyperlipidemia is
receptors. Important apolipoproteins are: as follows:
• A-I: Activates lecithin-cholesterol acyl transferase • Type I: Increased chylomicrons
(LCAT) • Type IIA: Increased β lipoproteins (LDL)
• B-100: Ligand for LDL receptors • Type IIB: Increased β and pre-β lipoproteins (LDL,
• C-II: Activates lipoprotein lipase VLDL)
• E: Ligand for chylomicron remnant receptor in liver • Type III: Broad β lipoproteins (IDL)
• Type IV: Increased pre-β lipoproteins (VLDL)
CLASSIFICATION OF LIPOPROTEIN • Type V: Increased chylomicrons and pre-β lipo-
DISORDERS proteins (chylomicrons, VLDL)
Secondary lipoprotein diseases arise from an
In clinical practice, lipoprotein disorders are classified underlying cause such as diabetes mellitus, alcohol abuse,
as being primary (inherited disorders) or secondary hypothyroidism, nephrotic syndrome, renal failure, and
(acquired disorders). Primary lipoprotein disorders are biliary cirrhosis of liver.

Table 5.2: Frederickson’s classification of primary lipoprotein disorders

Types Prevalence Appearance of plasma Increased particles Blood lipids


(fasting)

I Rarest Creamy layer at the top Chylomicron Marked elevation of triglycerides


IIA Common; 1:500 Clear (orange-yellow tint) Low density lipoprotein Increased total cholesterol
IIB Common; 1:300 Clear to slightly turbid Low density lipoprotein, Increased triglycerides and total
Very low density lipoprotein cholesterol
III Uncommon Thin creamy layer at the top; Intermediate density Increased triglycerides
turbid to opaque plasma lipoprotein and total cholesterol
IV Common; 1:500 Turbid to opaque Very low density lipoprotein Increased triglycerides
V Uncommon Creamy layer at the top; Chylomicron, Very low Marked elevation of triglycerides
plasma turbid to opaque density lipoprotein

In routine clinical practice lipid abnormalities are identified by standard lipid assays.

tahir99 - UnitedVRG
vip.persianss.ir
Disorders of Lipids and Biochemical Cardiac Markers 73

Fig. 5.5: Classification of lipoproteins by agarose gel


electrophoresis

The main risk factors for coronary artery disease are


age (men > 45 years, women > 55 years), dyslipidemia,
family history of premature coronary heart disease,
hypertension, and cigarette smoking. Lipid abnor-
malities associated with atherosclerosis and increased
risks of coronary artery disease are (i) elevated LDL
cholesterol, (ii) low HDL-cholesterol, and (iii) elevated
triglycerides.

LABORATORY TESTS FOR


LIPOPROTEIN DISORDERS
Collection of sample: Sample for lipid analysis should Fig. 5.6: Approach to the evaluation of a lipid disorder
be collected in dry EDTA anticoagulant (1 mg/ml). For
estimation of triglycerides and lipoproteins, a 12-hour
fasting sample is necessary. Triglycerides and LDL are It is recommended by National Cholesterol Education
affected by recent ingestion of food. Patient should be Program that a fasting lipid profile (consisting of
on the usual diet for 2-3 weeks before analysis. Lipid triglycerides, total cholesterol, HDL, and LDL) be carried
analysis should not be performed during acute illness out every 5 years beginning at the age of 20 years.
and should be deferred for 3 months after major illness. Desirable and high risk levels of serum lipids are shown
Drugs affecting lipid levels should be avoided like in Table 5.3.
steroids, oral contraceptives, etc.
(1) Total cholesterol: Causes of elevated serum
Approach to the evaluation of lipid disorder is shown
cholesterol are listed in Table 5.4.
in Figure 5.6.
Identification of a lipid disorder: Following investi- (2) Serum triglycerides: Hypertriglyceridemia is a risk
gations on a fasting blood sample are usually adequate factor for coronary heart disease, but less significant than
for identifying lipid abnormalities in majority of cases: total cholesterol and lipoproteins. Patients with serum
• Total cholesterol triglycerides >200 mg/dl have risk of atherosclerosis, and
• Triglycerides >1000 mg/dl are at increased risk of acute pancreatitis.
• HDL-cholesterol Increase in triglyceride is often associated with low HDL.
• LDL-cholesterol Causes of hypertriglyceridemia are listed in Table 5.5.

Table 5.3: Guidelines of National Cholesterol Education Program Adult treatment Panel III (All values in mg/dl)

• Total cholesterol: Desirable: <200; Borderline high: 200-239; High: ≥ 240


• Triglycerides: Normal: <150; Borderline high:150-199; High:200-499; Very high: ≥ 500
• LDL: Optimal: <100; Near optimal: 100-129; Borderline high: 130-159; High: 160-189; Very high: ≥ 190
• HDL: Low: <40; High: ≥ 60.
74 Essentials of Clinical Pathology

Table 5.4: Causes of hypercholesterolemia

Primary Secondary

1. Type IIA and type IIB hyperlipidemia (Familial hypercholesterolemia) 1. Hypothyroidism


2. Common or polygenic hypercholesterolemia 2. Nephrotic syndrome
3. Familial apolipoprotein B100 defect 3. Cholestasis
4. Familial alpha lipoproteinemia 4. Drugs: Protease inhibitors

Table 5.5: Causes of hypertriglyceridemia

Primary Secondary

1. Type I hyperlipidemia 1. Obesity


2. Type V hyperlipidemia 2. Poorly controlled diabetes mellitus
3. Deficiency of lipoprotein lipase 3. Alcoholism
4. Deficiency of apolipoprotein C-II 4. Renal failure
5. Drugs: Estrogen, glucocorticoids

VLDL level can be derived from serum triglyceride measured directly or can be derived from Friedewald’s
level by the formula: equation as follows:
LDL cholesterol = Total cholesterol – (HDL cholesterol – VLDL)
Triglycerides
VLDL = Triglycerides
5 and VLDL =
5
(3) HDL-cholesterol: HDL contains 20-30% of total
Identification of cause of dyslipidemia: After the
serum cholesterol. Low HDL-cholesterol is a significant
risk factor for coronary artery disease even if total presence of dyslipidemia is established, it is necessary
to determine whether it is primary or secondary on the
cholesterol level is normal. HDL is involved in “reverse
basis of clinical features, family history, and laboratory
cholesterol transport” (i.e. from peripheral tissues to the
studies. Initially, the secondary causes should be ruled
liver where it is excreted in bile) and thus decreases
out. Laboratory studies for identification of the secondary
cholesterol accumulated in blood vessel walls. Therefore,
nature of the disorder are blood glucose (diabetes
it is called as cardioprotective cholesterol. Concentration
of HDL-cholesterol is inversely related with the risk of mellitus), liver function tests (biliary cirrhosis), thyroid
function tests, and plasma and urine albumin (nephrotic
atherosclerotic coronary artery disease. HDL-cholesterol
syndrome).
<40 mg/dl is an important risk factor for coronary artery
disease (positive risk factor), while level >60 mg/dl is
BIOCHEMICAL CARDIAC MARKERS
cardioprotective (negative risk factor).
(4) LDL-cholesterol: LDL contains about 60% of total Acute Coronary Syndrome
serum cholesterol. High LDL-cholesterol is a strong risk The term acute coronary syndrome comprises of condi-
factor for atherosclerotic heart disease, and is the major tions characterized by acute myocardial ischemia and
atherogenic lipoprotein. It is the primary lipoprotein that includes (i) unstable angina, (ii) non-ST segment
mediates atherosclerotic heart disease, and is the primary elevation myocardial infarction (NSTEMI) and (iii) ST
target of lipid-lowering therapy. High LDL levels are segment elevation myocardial infarction (STEMI)
associated with obesity, high carbohydrate intake, (Table 5.6). The basic pathogenetic mechanism is
diabetes mellitus, lack of exercise, smoking, and some atherosclerosis of a coronary artery; acute coronary
drugs. Intensive therapy to lower LDL cholesterol slows syndrome results from rupture or erosion of an athero-
the progression of atherosclerosis, reduces coronary matous plaque with subsequent superimposition of
events, and decreases mortality. LDL-cholesterol can be thrombosis (Fig. 5.7). Clinical presentation is decided by
Disorders of Lipids and Biochemical Cardiac Markers 75

Table 5.6: Characteristics of acute coronary syndromes


Parameter Unstable angina (UA) Non-ST segment elevation ST segment elevation myocardial
myocardial infarction infarction (STEMI)
(NSTEMI)

1. Clinical features Three main presentations: Similar to UA or STEMI Pain similar to angina but more severe
1. New angina of severe onset, and persistent; not completely relieved
2. Angina at rest, or by rest or nitroglycerin; nausea, sense
3. Recent increase in frequency, of apprehension, and sweating;
duration and severity of angina asymptomatic in 25%
2. ECG ST depression and/or T wave ST depression and/or ST segment elevation followed by
inversion OR Normal T wave inversion OR appearance of Q wave and T
Normal wave inversion
3. Biomarkers of Not raised Raised Raised
myocardial
injury in blood

Fig. 5.7: Pathogenesis and classification of acute coronary syndrome. Acute coronary syndrome
results from acute reduction of myocardial blood supply due to disruption of atheromatous plaque

the degree of ischemia, amount of collateral circulation, The term acute coronary syndrome is coined to
myocardial oxygen demand, and certain other patient- distinguish between chronic stable angina from unstable
specific determinants. As the condition is life-threatening, angina and acute myocardial infarction.
early recognition of acute coronary syndrome is essential Majority of patients with acute coronary syndrome
so that proper and timely treatment can be instituted. have prior history of effort angina or coronary artery
The essential parameters for diagnosis are clinical disease. The usual clinical manifestation of myocardial
features, electrocardiographic changes, and detection of ischemia is chest pain. The pain is typically located in
biochemical markers released in blood from myocardial centre or left side of chest, and variously described as
damage. pressure, squeezing, constriction, crushing, tightness or
76 Essentials of Clinical Pathology

shown in Figure 5.8) but is more severe and persistent


(>30 minutes), and not readily relieved by rest or
nitroglycerin. In myocardial infarction pain is often
accompanied by other features such as sweating, nausea,
vomiting, breathlessness, and palpitations. Acute
myocardial infarction may be clinically silent (in 25%
cases, especially those with diabetes or hypertension).
Unstable angina can present with (i) new angina of
severe onset, (ii) angina at rest, or (iii) recent increase in
frequency or pattern of angina.
Previously, diagnosis of acute myocardial infarction
required any two of the following criteria (World Health
Fig. 5.8: Myocardial infarction about 1-2 days old showing Organization, 1974):
disappearance of nuclei of myocardial fibers, contraction bands, • Symptoms of myocardial ischemia, i.e. severe and
and beginning of acute inflammation. Rise in CK-MB enzyme prolonged chest pain,
occurs at this time • Unequivocal changes consistent with acute
myocardial infarction on electrocardiogram
heaviness. The pain radiates to left arm, shoulder, neck, (development of Q wave)
and jaw. It develops after exertion, meals, or emotional • Elevation of cardiac enzymes in blood
stress. In angina pectoris, pain is relieved by rest and A 12-lead electrocardiogram (ECG) is an important
nitroglycerin. Pain is similar in acute myocardial test for diagnosis and should be obtained as soon as
infarction (necrosis of myocardium due to ischemia as possible after presentation (Fig. 5.9). This is because the

Fig. 5.9: (1) Normal electrocardiogram pattern; (2) to (6): Sequential electrocardiographic changes after acute myocardial
infarction. Initially T wave becomes tall, peaked, and pointed (first few minutes) and there is ST segment elevation. T wave
inversion occurs after a few hours and there is development of an abnormal Q wave. After some duration, ST segment returns
to normal and T wave becomes normal. Q wave changes, however, persist.
Disorders of Lipids and Biochemical Cardiac Markers 77

test is noninvasive, inexpensive and rapid. The test is


useful for diagnosis, prognosis, and monitoring of
myocardial infarction. General ECG changes of
myocardial ischemia are ST elevation/depression and
deep symmetric T wave inversion. Persistent elevation
of ST segment differentiates STEMI from unstable angina
and NSTEMI. Normal appearing ECG does not exclude
cardiac ischemia or myocardial infarction.
ECG changes like ST segment elevation or depression
and T wave inversion are not sufficient on their own for
diagnosis of myocardial infarction. Revised criteria for
diagnosis of myocardial infarction (acute, evolving, or
recent) have been proposed by European Society of
Cardiology (ESC) and American Society of Cardiology
(ACC) (2000) (Box 5.1).

Box 5.1: Revised criteria for diagnosis of myocardial


infarction (ESC/ACC, 2000)
Any one of the following is satisfactory for diagnosis of Fig. 5.10: Cardiac muscle cell. Appearance of biochemical
acute/evolving/recent myocardial infarction: markers and their kinetics in bloodstream depend on their (i)
1. Typical myocardial necrosis-associated rise and fall intracellular location (whether free or bound to other organelles),
of troponin or CK-MB PLUS atleast one of the (ii) molecular weight, and (iii) rate of elimination from blood
following:
• Symptoms of ischemia
• Pathologic Q wave on ECG Table 5.7: Biochemical markers of myocardial injury
• ST segment elevation or depression on ECG
(indicative of ischemia) • Creatine kinase (CK)
• Coronary artery intervention (e.g. coronary – Total CK
angioplasty) – Isoenzymes
2. Pathologic features of an acute myocardial infarction – CK-MB (activity)
– CK-MB (mass)*
• Aspartate aminotransferase (AST) activity
Necrosis of myocardial cells leads to the release of
• Lactate dehydrogenase (LDH)
intracellular macromolecules from the cells into the – LDH activity
blood- stream. Detection of significant amounts of these – Isoenzymes
biochemical markers in blood under appropriate clinical • Cardiac troponins (cTn)*
setting can allow diagnosis of myocardial infarction to – cTnT
be made. Measurement of these markers can also – cTnI
distinguish between unstable angina and acute • Myoglobin*
myocardial infarction.
*Currently recommended markers
Biochemical Cardiac Marker Studies
troponin (a definitive marker) are the recommended
Myocardial cell necrosis leads to membrane damage and markers for diagnosis of MI; if cardiac troponin is not
leakage of cell contents into the bloodstream. This forms available, CK-MB (mass assay) is the next best alterna-
the basis for measurement of biochemical markers of tive.
myocardial injury in blood (Fig. 5.10). Various Usefulness of cardiac markers depends on time of
biochemical markers for myocardial injury are listed in specimen collection after myocardial infarction. Use of
Table 5.7. combination of markers and serial changes are more
Previously, total creatine kinase, LD, and AST were helpful (Table 5.8).
used for diagnosis of MI. However, due to their low An ideal cardiac marker should be highly sensitive,
specificity and due to the availability of more specific highly specific, should help to improve patient outcome,
markers, these enzymes are now rarely measured. and be cost-effective. Such a marker is currently
Currently, myoglobin (an early marker) and cardiac unavailable.
78 Essentials of Clinical Pathology

Table 5.8: Timeline of cardiac markers after acute MB is the most cardiac-specific CK isoenzyme, and it is
myocardial infarction recommended to measure CK-MB mass.
Markers Time for Peak Return to Total serum CK and CK-MB are always elevated
detection normal following MI. However, serum CK and CK-MB can arise
from tissues other than heart. Following MI, CK-MB rises
Myoglobin 1-3 hr 6-9 hr 1 day within 3-6 hours after the onset of symptoms, peaks
CK-MB 3-6 hr 12-24 hr 2-3 days within 12-24 hours, and returns to normal level by 48-72
hours.
Troponin 4-8 hr 12-24 hr 5-10 days
Relative index or RI ([CK-MB/total CK × 100) is used
to distinguish cardiac from skeletal muscle damage; RI
Role of biochemical cardiac markers in acute coronary above 5% is highly suggestive of acute myocardial
syndrome: infarction.
• To confirm or exclude the diagnosis of acute It is recommended to obtain sequential samples (one
myocardial infarction in cases with sudden onset of at presentation and subsequently at 8-hour intervals for
chest pain: In the presence of typical history and ECG 24 hours).
findings consistent with acute MI, measurement of Myoglobin: Myoglobin is the oxygen-binding low
biochemical markers does not provide additional molecular weight protein of cardiac and skeletal muscle
information for initial management. These patients cells. Myoglobin rises early after MI (1-3 hours) and is
need urgent treatment in the form of thrombolytic currently the earliest marker. Myoglobin of cardiac
therapy or angioplasty or both. If diagnosis is muscle cannot be distinguished from that of skeletal
uncertain from clinical features and ECG in an muscle. Myoglobin levels are raised following MI, open
emergency setting, biochemical markers are helpful
heart surgery, muscle injury, muscle dystrophy, renal
in ruling out myocardial infarction.
failure, shock, and trauma. Thus, although myoglobin
Serial determinations (at admission, 6-9 hours, and
rises early following MI, it is not cardiac-specific.
12-24 hours) are recommended to rule in or rule out
diagnosis of acute myocardial infarction. Use of CK-MB However, non-elevation of myoglobin (in two sequential
(mass) and cardiac troponin is recommended for samples 2-4 hours apart) is helpful for exclusion of early
diagnosis. MI in patients presenting with chest pain at emergency
• Detection of old (by some days) myocardial infarction department.
by cardiac troponin
Troponins (Tn): Cardiac troponin T (cTnT) and cardiac
• Diagnosis of reinfarction (CK-MB mass assay)
troponin I (cTnI) are the most sensitive and specific of
• To assess effectiveness of immediate reperfusion
the available markers of myocardial necrosis and are
therapy (thrombolysis or percutaneous coronary
intervention) in STEMI. considered ideal markers for definitive diagnosis (either
• Risk stratification to determine the likelihood of acute cTnT or cTnI). Troponins regulate the interaction of actin
coronary syndrome and myosin filaments during myocardial contraction.
Following MI, troponins appear in blood at about the
Creatine kinase: Highest activity of CK is present in
same time as CK-MB. If troponins are elevated atleast 12
striated muscle, brain, and heart.
or more hours following onset of chest pain, their
Causes of increased CK are as follows: diagnostic sensitivity is 100%. TnI is more cardio-specific
• Disorders of skeletal muscle: Trauma, intramuscular as it is found only in heart muscle. It is not elevated
injection, vigorous exercise, dermatomyositis, mus-
following skeletal muscle injury. Following myocardial
cular dystrophy
damage, TnI rises 4-8 hours the onset of chest pain, peaks
• Disorders of heart: Myocardial infarction, myocarditis
• Disorders of central nervous sytem: Cerebrovascular within 12-24 hours, and remains elevated for 7-10 days.
accident, head injury, generalized convulsions Development of assays for TnI and TnT represent a major
• Disorders of thyroid: Hypothyroidism advance in the diagnosis of MI. As troponins remain
CK-BB, CK-MB, and CK-MM are the isoenzymes of elevated for 7-10 days, they are useful in cases presenting
CK. CK-BB predominates in brain, CK-MB in cardiac late. If onset of chest pain is 9-12 hours before admission,
muscle, while CK-MM in skeletal muscle and heart. CK- only troponin needs to be measured.
Disorders of Lipids and Biochemical Cardiac Markers 79

REFERENCE RANGES 2. Bock JL. Test strategies for the detection of myocardial
damage. Clin Lab Med 2002;22:357-75.
• Lipid profile: 3. Crook MA. Clinical chemistry and metabolic medicine
– Serum cholesterol: Desirable level: <200 mg/dl 7th Ed. London. Edward Arnold (Publishers) Ltd. 2006.
– Serum triglycerides: Desirable level: <150 mg/ 4. Eaton CB. Hyperlipidemia. Prim Care Clin Office Pract
2005;32:1027-55.
dl
5. Executive Summary of The Third Report of The
– HDL cholesterol: >60 mg/dl
National Cholesterol Education Program (NCEP).
– LDL cholesterol: <130 mg/dl Expert Panel on Detection, Evaluation, and Treatment
– LDL/HDL ratio: 0.5-3.0 of High Blood Cholesterol in Adults (Adult Treatment
• Biochemical cardiac markers: Panel III). JAMA 2001;285:2486-97.
– CK-MB: <5% or <10 μg/L 6. Koolman J, Roehm KH. Color Atlas of Biochemistry,
– Cardiac Troponin T: <0.1 μg/L 2nd Ed. Stuttgart. Thieme 2005.
– Myoglobin: < 90 μg/L 7. McDermott MT. Endocrine secrets. 4th Ed.
Philadelphia: Mosby, 2005.
8. Senger AK, Jaffe AS. The use of biomarkers for the
CRITICAL VALUES evaluation and treatment of patients with acute
• CK-MB: >5% or >10 μg/L coronary syndromes. Med Clin N Am 2007;91:657-81.
9. The Joint European Society of Cardiology/American
• Cardiac Troponin T: >0.1 μg/L
College of Cardiology Committee: Myocardial infarc-
• Myoglobin: >110 μg/L tion redefined-A consensus document of The Joint
European Society of Cardiology/American College of
BIBLIOGRAPHY Cardiology Committee for the redefinition of myo-
1. Achar SA, Kundu S, NorcrossWA. Diagnosis of acute cardial infarction. Eur Heart J 2000;21:1502-13.
coronary syndrome. Am Fam Physician 2005;72: 10. Tiyyagura SR, Smith DA. Standard lipid profile. Clin
119-26. Lab Med 2006;26:707-32.
80 Essentials of Clinical Pathology
6

Examination of
Cerebrospinal Fluid

Cerebrospinal fluid (CSF) is a clear, colorless fluid formed • Chloride: 120-130 mEq/L (20 mEq/L more than
in the ventricles of the brain mainly by choroid plexus serum level)
(meshwork of tiny small blood vessels in lateral third • Bilirubin: Absent.
and fourth ventricles). It is mainly an ultrafiltrate of
plasma. CSF is contained within the cerebral ventricles, FUNCTIONS OF CEREBROSPINAL FLUID
the spinal canal and the subarachnoid space (space
between arachnoid externally and pia mater internally) 1. Protection of brain and spinal cord from injury by
surrounding the brain and spinal cord (Fig. 6.1). CSF is acting as a shock absorber.
reabsorbed into the blood through the arachnoid villi of 2. To serve as a medium between blood and brain for
dural venous sinuses. supply of nutrients to and removal of waste products
from brain.
COMPOSITION OF NORMAL COLLECTION OF CEREBROSPINAL FLUID
CEREBROSPINAL FLUID IN ADULTS
Some diseases produce characteristic alterations in
• Total volume: 100-150 ml (10-60 ml in the newborn) composition of CSF, thus providing the basis for
• Opening pressure: 60-180 mm of water (10-100 mm examination of CSF. Lumbar puncture or LP (first
in infants and young children) performed by Quincke in 1891) is the procedure whereby
• Appearance: Clear and colorless with no clots a sample of CSF is obtained. Spinal or LP needle is passed
(viscosity similar to water) between 3rd and 4th or between 4th and 5th lumbar
• Cells: vertebra (L3-L4 or L4-L5) and CSF is obtained from the
– Adults: 0-5 cells/cmm subarachnoid space (Fig. 6.2). LP is carried out at these
– Infants: 0-30 cells/cmm
– 1-4 years: 0-20 cells/cmm
– 5-18 years: 0-10 cells/cmm
• Glucose: 45-80 mg/dl. (Normally CSF glucose is 60%
or 2/3rds of blood glucose)
• Proteins: 15-45 mg/dl. (Normally CSF proteins are
1% of plasma proteins).
• Oligoclonal bands: Negative

Fig. 6.2: Site of lumbar puncture. Structures through which


needle passes are skin/superficial fascia, ligaments, epidural
space, dura mater, subdural space, arachnoid mater and
Fig. 6.1: Cranial meninges subarachnoid space
Examination of Cerebrospinal Fluid 81

Fig. 6.3: Lumbar puncture needle

levels to prevent injury to the spinal cord (spinal cord


ends at about T12, below which are nerve roots or cauda
equina).
Patient is in a side-lying (lateral recumbent) position
with his back absolutely vertical and at the edge of the
bed, with the knees drawn up and the head flexed onto
his chest. This position increases the space between the
lumbar vertebrae. Alternately, patient may be in a sitting
position.
The selected site is thoroughly disinfected with
povidone-iodine or chorhexidine-containing solution,
covered with sterile drapes, and after injecting a local
anesthetic, a sterile lumbar puncture needle, preferably
22 gauge (Fig. 6.3), is inserted slowly. There is an
increased resistance as the needle passes through the
spinal ligaments and dura mater. As the needle enters
the subarachnoid space, a sudden 'give' (or loss of Fig. 6.4: Laboratory tests for evaluation of CSF in
resistance) is felt. The stylet is withdrawn slowly. When different tubes
CSF drops appear, a pre-assembled manometer is
attached to the needle and the opening pressure is • Meningeal involvement by leukemia or
recorded. To measure opening pressure, patient should malignancy
be in the lateral recumbent position. • Subarachnoid hemorrhage (if CT scan is not
With the three-way stopcock in appropriate position, available)
CSF is collected in sterile plain tubes as follows (Fig. 6.4): • Inflammatory diseases, e.g. multiple sclerosis (for
• Tube 1: Chemistry (glucose and protein) and diagnostic gammaglobulin findings), Guillain-
serology Barré syndrome
• Tube 2: Microbiology (Gram's staining, bacterial • Neoplasms of central nervous system (CNS).
culture, and sensitivity)
• Tube 3: Hematology (Total cell count and Indications for emergency lumbar puncture are
differential count) shown in Box 6.1.
• Tube 4: Cytology, special studies. 2. To reduce CSF pressure in benign intracranial
Typically, 3-5 ml of CSF is collected. After CSF hypertension (pseudotumor cerebri)
collection, the closing pressure is recorded (if the opening 3. Administration of medications:
pressure was high). • Anesthetic agents
The needle is withdrawn after replacing the stylet, • Antibiotics (e.g. amphotericin B in fungal menin-
and a sterile dressing is applied. Before or immediately gitis)
after LP, venous blood should be obtained for concurrent • Anticancer drugs (e.g. methotrexate in acute
determination of blood glucose. lymphoblastic leukemia)
4. Introduction of radiographic contrast media for
INDICATIONS FOR LUMBAR PUNCTURE myelography
1. Examination of CSF for diagnosis in suspected cases 5. For Queckenstedt's test: If opening CSF pressure is
of: normal and clinically subarachnoid block or a tumor
• Central nervous system infections, especially or a sinus thrombosis is suspected, then
meningitis (inflammation of leptomeninges) and Queckenstedt's test may be preformed. However, the
encephalitis test is contra-indicated if CSF pressure is raised since
82 Essentials of Clinical Pathology

it can precipitate herniation of brain. There is a direct tumor, subdural hematoma, epidural abscess): These
correlation between CSF pressure and pressure in patients usually have headache, altered pupillary
jugular vein (because of continuity with dural venous response, absent Doll's eye reflex, abnormal
sinuses in which arachnoid villi project). In
respiratory pattern, papilledema, bradycardia,
Queckenstedt's test, both jugular veins are
hypertension, and decerebrate or decorticate
compressed and then released, and subsequent
changes in CSF pressure are observed. Normally, posturing. Lumbar puncture in such cases can lead
upon compression of jugular veins, CSF pressure to herniation of brain. If a mass lesion is clinically
rapidly rises and with the release of pressure on suspected, cranial computerized tomography (CT) or
jugular veins, CSF pressure rapidly falls. In the magnetic resonance imaging (MRI) should be
presence of a spinal block, there is no rise of CSF obtained first.
pressure with jugular vein compression (or a pressure 2. Cardiorespiratory compromise
rise is low or delayed). This test is positive in about
3. Bleeding diathesis that has not been corrected
80% of patients with cord compression. With the
4. Local infection at the site of lumbar puncture
advent of myelography, Queckenstedt's test is rarely
performed. LABORATORY EXAMINATION OF
Box 6.1: Indications for emergency lumbar puncture CEREBROSPINAL FLUID
• Suspected meningitis After collection, specimen of CSF should be transported
• Suspected subarachnoid hemorrhage immediately to the laboratory and examined without
• Suspected meningeal involvement by leukemia delay. This is because (i) cells disintegrate rapidly, and
(ii) reduction of glucose level occurs due to glycolysis.
COMPLICATIONS OF LUMBAR PUNCTURE At the latest, CSF should be examined within 1 hour of
1. Post-puncture headache: This is the most common collection, and CSF cell counts are always done within
side effect and results from leakage of CSF from 30-60 minutes of collection. Glass tubes should not be
puncture site at a rate faster than the rate of CSF used for collection since cell adherence to glass reduces
production. With the use of a large bore needle, the cell count. Specimen for bacterial culture should not
greater CSF leak occurs. Use of a smaller bore needle be refrigerated as fastidious organisms (Hemophilus
(22 G) for LP and keeping the patient flat after the influenzae, Neisseria meningitidis) do not survive in the cold
procedure for 2-3 hours reduces the risk of headache. temperature.
2. Introduction of infection in spinal canal if aseptic CSF chemical examination results should always be
precautions are not observed, if septicemia is present, compared with those in plasma since any change in
or if infection is present at the site of LP. plasma is reflected in CSF.
3. Subdural hematoma with resultant neurologic deficit
Examination of CSF includes:
in patients with bleeding diathesis.
1. Opening pressure
4. Failure to obtain CSF (dry tap) which may be due to
the incorrect positioning of the patient or spinal block. 2. Appearance
5. Herniation of brain through tentorium (uncus of 3. Total and differential cell counts
temporal lobe) or foramen magnum (cerebellar 4. Chemical examination
tonsils), if intracranial pressure is high. This can 5. Microbiological examination
damage the brainstem. This risk has led to the 6. Special investigations
reduced use of lumbar puncture over the last few
years. 1. Opening Pressure
6. Subarachnoidal epidermal cyst (due to traumatic After attaching the manometer to the hub of the spinal
implantation of a skin plug in subarachnoid space) needle, patient's legs should be gently extended and neck
may develop after a few years if LP is preformed returned to neutral position. The CSF pressure is then
without a stylet. measured. CSF pressure is directly related to jugular and
vertebral venous pressures. Patient should be relaxed
CONTRAINDICATIONS TO LUMBAR PUNCTURE since tension, straining, or breath-holding will increase
1. Raised intracranial pressure due to a space- the CSF pressure, while hyperventilation will lower the
occupying lesion (e.g. brain abscess, posterior fossa opening pressure.
Examination of Cerebrospinal Fluid 83

Normal opening pressure of CSF is:


• 60-180 mm of water in adults in lateral recumbent
position
• 10-100 mm of water in children less than 8 years
Causes of increased CSF pressure:
• Tense and anxious patient
• Intracranial mass lesion (e.g. neoplasm, abscess,
hemorrhage)
• Meningitis
• Cerebral edema
• Subarachnoid hemorrhage
• Congestive cardiac failure
• Benign intracranial hypertension (pseudotumor
cerebri).
Causes of decreased CSF pressure: Fig. 6.5: Appearance of CSF. (1) Normal CSF is clear and
• Leakage of spinal fluid following trauma or previous transparent. (2) Bloody CSF (either due to traumatic LP or
lumbar puncture subarachnoid haemorrhage). (3) After centrifugation, bloody
• Complete spinal block (obstruction of spinal sub- CSF due to traumatic LP shows clear supernatant, while that
arachnoid space due to tumor, abscess, adhesions, due to subarachnoid haemorrhage shows, (4) yellowish
herniated intervertebral disk). supernatant (xanthochromia) (5) Turbid CSF
A large difference between opening and closing
pressures usually indicates presence of a partial or – Leukocytes >200 cells/cmm
complete spinal block. – Red cells >400 cells/cmm
If opening CSF pressure is >200 mm, no more than 1- – Microorganisms like bacteria, fungi, or amebae
2 ml of CSF should be removed. – Radiographic contrast media
– Aspiration of epidural fat during LP
2. Gross Appearance of Cerebrospinal Fluid – Raised proteins.
Normal CSF is clear and colorless like distilled water, • Blood-mixed CSF: Blood-stained CSF may result
and does not clot. Abnormal CSF may appear turbid, from traumatic tap (due to injury to venous plexus in
blood-mixed, xanthochromic, or viscous (Fig. 6.5). Clot spinal wall) or subarachnoid hemorrhage. Distinction
formation in CSF is abnormal and indicates increased of traumatic tap from subarachnoid hemorrhage is
proteins. vitally important. Differences between the two are
• Turbid CSF may be due to the presence of: given in Table 6.1.

Table 6.1: Differences between traumatic lumbar puncture and subarachnoid hemorrhage

Cerebrospinal fluid finding Traumatic lumbar puncture Subarachnoid hemorrhage

1. Gross appearance Blood more in initial tubes as Blood uniform in all tubes; Blood does not
compared to later tubes; clot on standing
Blood clots on standing.
2. Supernatant after centrifugation Clear Pink or yellow (xanthochromia); yellow
within 1 hour of collection xanthochromia develops 12 hours after
hemorrhage
3. Microscopy Progressive decrease of red cell Red cell counts uniform in all tubes;
counts in later tubes Hemosiderin-laden macrophages
present
4. Latex agglutination test for Negative Positive
D-dimer*
5. Cerebrospinal fluid pressure Normal Increased
6. Cerebrospinal fluid protein Normal Increased

*D-dimer: A cross-linked fibrin degradation product


84 Essentials of Clinical Pathology

• Xanthochromia: This refers to yellow discoloration 3. Cell Counts in Cerebrospinal Fluid


of CSF. For its detection, CSF is centrifuged and the
(1) Total leukocyte count: Cell count on CSF is done
supernatant is compared with another tube of same manually on undiluted sample in a counting chamber.
size filled with distilled water. Causes of Total leukocyte count increases in various disorders
xanthochromia are: and along with differential count provides important
– Subarachnoid hemorrhage (12 hours after diagnostic information. An increase in cell count in CSF
bleeding episode): In subarachnoid hemorrhage, is called as pleocytosis.
bleeding occurs in subarachnoid space usually It is essential to do microscopic examination of all
due to rupture of a cerebral aneurysm. Patient CSF samples since white blood cell (WBC) count upto
presents with severe bursting headache of sudden 200/cmm and red cell count upto 400/cmm are associated
onset in occipital region that may be followed by with clear appearance of CSF.
loss of consciousness. Red blood cells in CSF are For correct results:
hemolyzed with release of oxyhemoglobin. About • Cell count should be done as soon as possible after
a week after bleeding, macrophages and other collection of CSF since cellular disintegration occurs
cells of leptomeninges convert oxyhemoglobin to rapidly. Cells also adhere to the walls of the glass
bilirubin. This produces yellowish discoloration tubes.
of CSF supernatant (See Fig. 6.5). • CSF specimen collected in tube 3 should be used.
The investigation of choice, if available, for • No dilution of CSF is usually required. A diluent
should be used only if CSF is cloudy and likely to
diagnosis of subarachnoid hemorrhage is CT scan
contain increased leukocytes.
to detect blood in basal cisterns. When CT scan is
negative or equivocal and xanthochromia cannot Method:
be appreciated visually, spectroscopic i. CSF sample should be properly mixed. If CSF is
examination of CSF is helpful for diagnosis of clear, it is not diluted. If CSF appears cloudy or
subarachnoid hemorrhage. It will show turbid, 1:20 dilution is made using 0.05 ml of CSF
absorption peaks of oxyhemoglobin and bilirubin. and 0.95 ml of Turk solution. (Composition of Turk
– Jaundice, when serum bilirubin is >6.0 mg/dl solution: Glacial acetic acid 4 ml, methylene blue
solution 10 drops, and distilled water to make 200
– CSF protein >150 mg/dl.
ml).
Froin's syndrome is a combination of xanthochromia,
ii. The counting chamber is covered with the coverslip
excess proteins in CSF, and spontaneous formation of a provided.
coagulum in CSF on standing. It results from complete iii. The counting chamber is filled with the fluid and
block of subarachnoid space. allowed to stand for 2 minutes for cells to settle.
• Other abnormal colors of CSF: These are: iv. For counting cells in CSF, Fuchs-Rosenthal counting
– Pink: Red cell lysis and hemoglobin breakdown chamber is preferred because its depth is twice that
– Brownish: Meningeal metastatic melanoma of improved Neubauer chamber. In this, cells are
– Orange: High carotene ingestion counted in 5 large squares (4 corner squares and
• Clot formation: Pellicle (thin membrane or scum on one central square).
the surface of CSF) or clot formation (after 10 minutes v. If undiluted CSF is used, total number of cells
of collection) indicates increased proteins (>150 mg/ counted in 5 squares represents total count per cmm
of CSF. If CSF is diluted, the number of cells counted
dl). It occurs in tuberculous meningitis (fine cobweb-
is multiplied by the dilution factor (i.e. 20).
like clot after 12-24 hours), purulent meningitis
(pellicle forms early followed by a large clot), spinal Causes of increased cell count in CSF:
block (complete clotting of CSF), and traumatic LP. • Meningitis and other infections of CNS
Clot formation does not occur in subarachnoid • Intracranial hemorrhage
hemorrhage. • Meningeal infiltration by malignancy
• Repeated lumbar punctures
• Thick viscous CSF: This is seen cryptococcal
• Injection of foreign substances (e.g. radiographic
meningitis, meningeal metastatic mucinous adeno- contrast media, drugs) in subarachnoid space.
carcinoma, severe meningitis, and release of nucleus • Multiple sclerosis
pulposus fluid in CSF due to needle injury to the Presence of blood in CSF due to traumatic tap or
intervertebral disk. subarachnoid hemorrhage artefactually raises the
Examination of Cerebrospinal Fluid 85

If CSF contains only a few cells, then it is centrifuged


at high speed (3000 g) for 10 minutes and a smear is made
from the sediment. If CSF contains many cells, then a
smear is made directly from the uncentrifuged sample.
After staining with a Romanowsky stain, smear is
examined under the microscope (Fig. 6.6).
Simple centrifugation of CSF often causes cell
breakage and distortion. Cytospin preparation with
cytocentrifuge (high speed centrifugation to concentrate
cells on a slide in a uniform monolayer) has been
recommended as it improves the cell yield and preserves
the cell morphology well.
In normal adults, differential count shows 70%
lymphocytes and 30% monocytes. In young children, a
higher proportion of monocytes (up to 70%) are present.
Table 6.2 shows causes of increase in different types of
leukocytes in CSF.
(3) Other cells: Apart from mature blood cells, CSF may
contain immature hematopoietic cells, tissue cells
Fig. 6.6: Cells in CSF: (1) Many neutrophils in bacterial
(ependymal cells, pia arachnoid mesothelial cells) and
meningitis, (2) lymphocytes seen in normal CSF in adults, (3)
Many red cells and a small lymphocyte (traumatic tap), (4)
malignant cells. CSF examination is commonly carried
Increased monocytes in CSF, (5) Neutrophils, lymphocytes, out in acute lymphoblastic leukemia to detect
and red cells in CSF, (6) Lining cells of pia and arachnoid involvement of CNS. Increased WBC count (>5/μl) with
in CSF, (7) Blast cells in CSF, (8) a malignant cell in CSF lymphoblasts is evidence of CNS involvement.

leucocyte count by 1 WBC per 1000 red cells. This 4. Chemical Examination of Cerebrospinal Fluid
correction factor should be used if patient's hemogram
is normal. If significant anemia or leukocytosis is present, Routine chemical examination of CSF consists of estima-
then leukocyte count in CSF should be corrected as tion of proteins and glucose. CSF from tube 1 is used for
follows: chemical examination.
Corrected WBC count in CSF = (1) Estimation of proteins in CSF: Normal CSF protein
WBC count in blood × level in adults is 15-45 mg/dl. An increase in CSF protein
Red cell count in CSF is a sensitive but non-specific indicator of CNS disease.
WBC count in CSF (cells/cmm) –
Red cell count in blood CSF proteins may be normal during early stages of
(2) Differential leukocyte count: This provides informa- meningitis. Significant elevation (>150 mg/dl) occurs in
tion about relative proportion of various leukocytes. bacterial meningitis.

Table 6.2: Differential cell count in CSF

Predominant neutrophils Predominant Mixed cell pattern Predominant


lymphocytes (neutrophils, lympho- eosinophils
cytes, monocytes)

1. Meningitis: bacterial, 1. Meningitis: viral, tuberculous 1. Tuberculous meningitis 1. Parasitic and fungal
early viral, fungal, 2. Incompletely treated bacterial 2. Fungal meningitis infections
early tuberculous meningitis 3. Chronic bacterial 2. Reaction to foreign
2. Subarachnoid hemorrhage 3. Cysticercosis, toxoplasmosis meningitis material (e.g. shunts)
3. Repeated lumbar punctures 4. Multiple sclerosis
4. Introduction of anticancer 5. Subacute sclerosing
drugs or contrast media in panencephalitis
subarachnoid space
5. Meningeal metastasis
86 Essentials of Clinical Pathology

There are various methods for estimation of CSF


proteins. Turbidimetric method using trichloroacetic acid
for precipitation of proteins is commonly used. In
principle, trichloroacetic acid, when added to CSF, causes
precipitation of proteins and a turbid solution is obtained.
Amount of turbidity is compared with the turbidity of a
known (standard) concentration of protein in a photo-
electric colorimeter.
If a sample is contaminated with blood while doing
the lumbar puncture (traumatic tap), false elevation of
proteins will occur. This can be corrected by deducting
1 mg/dl of protein for every 1000 red cells per cmm. For
this correction to be accurate, red cell count and proteins
should be estimated in the sample from the same tube of
CSF. Fig. 6.7: Pandy's test for globulins
If facilities for estimation of CSF proteins are not
vs. increased intrathecal synthesis of immunoglobulin
available in the laboratory, then Pandy's test for globulins
G) can be made from parameters shown in Table 6.3.
may be performed. In this test, CSF is added to saturated
Albumin is neither synthesized nor metabolized in CNS.
solution of phenol. If cloudiness develops immediately,
Therefore, increased CSF albumin/serum albumin ratio
it indicates presence of increased globulins and the test
indicates increased permeability of blood-brain barrier.
is reported as positive. If no cloudiness develops, the test
Immunoglobulin G (IgG) can be synthesized in CNS.
is reported as negative (Fig. 6.7).
Therefore, increased CSF IgG/serum IgG ratio indicates
Along with CSF proteins, it is necessary to simulta-
either increased permeability of blood-brain barrier or
neously measure serum proteins for proper increased intrathecal synthesis of IgG. Increased CSF
interpretation of results. IgG/albumin index indicates local IgG production.
CSF proteins are elevated in following conditions:
• Increased capillary permeability of blood-brain
barrier: Meningitis
• Mechanical obstruction to circulation of CSF
(causing increased fluid reabsorption due to stasis):
Spinal cord tumor
• Increased local (intrathecal) immunoglobulin (IgG)
production: Multiple sclerosis, neurosyphilis, sub-
acute sclerosing panencephalitis
• Both increased capillary permeability and increased
local immunoglobulin (IgG) production: Guillain-
Barré syndrome
• Hemorrhage in CSF: Traumatic tap, subarachnoid (2) Estimation of glucose in CSF: Normal CSF glucose
hemorrhage. is 2/3rds of blood glucose (CSF to blood glucose ratio is
Marked elevation (>500 mg/dl) is noted in complete 0.6). A sample for blood glucose should be drawn 1 hour
spinal block by a tumor, bacterial meningitis, and bloody before LP for comparison with CSF glucose. After
CSF. collection, CSF sample should be immediately processed
Differential diagnosis of elevated proteins in CSF for glucose estimation because falsely low result due to
(increased capillary permeability of blood-brain barrier glycolysis may occur.

Table 6.3: Differentiation of causes of elevated proteins in cerebrospinal fluid

CSF/Serum albumin ratio CSF/Serum IgG ratio CSF IgG/Albumin index Causes

1. Increased Increased Normal Increased permeability of blood-brain barrier


2. Normal Increased Increased Increased intrathecal synthesis of proteins
Examination of Cerebrospinal Fluid 87

CSF glucose is measured by glucose oxidase method.


Normal range is 45-80 mg/dl. CSF glucose <40 mg/dl is
abnormal.
CSF glucose is decreased due to utilization by bacteria
(pyogenic or tuberculous), leucocytes, or cancer cells in
CSF.
Decreased CSF glucose occurs in following
conditions:
• Acute bacterial meningitis
• Tuberculous meningitis
• Fungal meningitis Fig. 6.8: India ink preparation of CSF showing single and
• Meningeal involvement by malignant tumor budding yeast cells of Cryptococcus neoformans
(meningeal carcinomatosis) nervous system. It causes a fatal type of hemorrhagic
• Hypoglycemia meningoencephalitis. Candida albicans may be seen in
CSF glucose is normal in viral meningitis. unstained wet mount; it appears as oval budding forms
and as pseudohyphae.
5. Microbiological Examination
When cryptococcal meningitis is clinically suspected,
Microbiological tests which can be carried out on CSF the wet mount is examined by dark-field microscopy.
sample are— Alternatively, a drop of India ink is added to the drop of
• Direct wet mount of CSF: in suspected cases of sediment on a glass slide, a coverslip is placed, and
cryptococcosis, amebic meningoencephalitis, examined under the microscope using ×40 objective.
Candida infection, and trypanosomiasis Cryptococcus neoformans appears as spherical, budding
• Gram's smear: should be done if CSF is turbid and yeast forms, 2-10 μ in diameter and surrounded by a large
neutrophils are increased. unstained capsule (Fig. 6.8). Cryptococci can be
• Ziehl-Neelsen smear: if tuberculous meningitis is demonstrated by India ink preparation in 50% cases of
suspected. cryptococcal meningitis.
• Latex agglutination tests: for detection of bacterial
(b) Gram's smear: This must be done if CSF is purulent
and cryptococcal antigens.
and neutrophils are increased. Gram's smear is positive
• Serologic tests for syphilis
in 80% untreated cases and 60% partially treated cases
• Culture for bacteria and Mycobacterium tuberculosis
of bacterial meningitis. Therefore, absence of bacteria on
• Polymerase chain reaction for Mycobacterium
Gram's smear does not rule out bacterial infection.
tuberculosis and viruses.
CSF is centrifuged and a smear of the deposit is made
(a) Direct wet mount of CSF: One drop of CSF deposit on a glass slide. (If CSF is purulent, smear is made directly
(obtained after centrifugation) is placed on a glass slide, without centrifugation). Smear is air-dried, stained with
covered with a cover glass, and examined under the Gram's method, and examined under the oil-immersion
microscope with reduced illumination. Observe for lens for bacteria (Fig. 6.9). Bacteria which commonly
motile trypanosomes or sluggishly moving amebae cause meningitis are:
(Naegleria fowleri). N. fowleri is a free-living ameba in water • Meningococci: Gram-negative diplococci located
which enters through the nose and reaches central inside neutrophils.

Fig. 6.9: Gram stained smears of CSF showing (1) Neisseria meningitidis,
(2) Streptococcus pneumoniae, and (3) Haemophilus influenzae
88 Essentials of Clinical Pathology

• Pneumococci: Gram-positive diplococci surrounded endotoxin. Endotoxin is produced by gram-negative


by an unstained capsule. bacteria like N. meningitides, H. influenzae type b, E. coli,
• Hemophilus influenzae: Gram-negative coccobacilli. and Pseudomonas. This test is particularly useful as a rapid
• Escherichia coli: Gram-negative rods. test in newborns in whom these infections are common.
In all the above types of meninigitis, the (f) Serologic tests for syphilis: If fluorescent treponemal
accompanying cells are polymorphonuclear neutrophils. antibody with absorption (FTA-ABS) test is positive in
Detection of typical bacteria on Gram-staining serum, Venereal Disease Research Laboratory (VDRL)
suggests the etiologic agent of meningitis. However, test should be done on CSF if neurosyphilis is suspected.
definitive diagnosis requires culture of CSF. VDRL test is highly specific but lacks sensitivity.
There is an association between age of the patient and Therefore, a positive test rules in but does not rule out
the causative organism of meningitis (Box 6.2). the diagnosis of neurosyphilis. Other serological tests for
syphilis are not suitable for diagnosis of neurosyphilis
(c) Ziehl-Neelsen staining for Mycobacterium tuber-
in CSF.
culosis: In tuberculous meningitis, number of tubercle
bacilli is usually low in CSF. Ziehl-Neelsen or AFB Combination of positive FTA-ABS test in serum and
staining is not a very sensitive method for detection of reactive VDRL test in CSF is diagnostic of active
M. tuberculosis in CSF. AFB smears are negative in about neurosyphilis
70% of cases of tuberculous meningitis. Fluorescent
(g) CSF culture: Culture of CSF is indicated if bacteria
auramine stains have better sensitivity than Ziehl-
are seen on Gram-stained smear, or leukocytes or
Neelsen stain.
proteins are increased. Culture remains the gold standard
for diagnois of bacterial meningitis. For culture, CSF
Box 6.2: Association between age and organisms
collected in tube 2 is used. Sensitivity of culture for
causing meningitis
identification of bacteria is about 90%. In incompletely
• 0 to 6 months: Group B streptococci, Escherichia treated cases, sensitivity is less. Usually CSF sample is
coli, Listeria monocytogenes inoculated on chocolate (heated blood) agar and blood
• 6 months to 6 years: Streptococcus pneumoniae, agar. In newborn infants, sample is also inoculated on
Neisseria meningitidis, Hemophilus influenzae type
McConkey's agar.
B, Enteroviruses
In tuberculous meningitis, culture for M. tuberculosis
• 6 to 60 years: Neisseria meningitides, Streptococcus
pneumoniae, Enteroviruses, herpes simplex virus is positive in about 56% of cases. If larger volume of CSF
• >60 years: Streptococcus pneumoniae, gram- is used (i.e. 10 ml) for inoculation, sensitivity for detection
negative bacilli, Listeria monocytogenes increases.
In cryptococcal meningitis, culture is positive in 95%
of cases.
(d) Latex agglutination tests: Latex agglutination tests
for bacterial antigens are available commercially and are (h) Polymerase chain reaction (PCR): PCR is a highly
sensitive, rapid, and simple to perform. Currently specific and sensitive tool for diagnosis of infections of
available tests can detect N. meningitidis (groups A, B, C, CNS. It uses probes to detect genes specific for the
Y, and W135), H. influenzae (capsular type B), S. infecting organism. The test is rapid and requires only a
pneumoniae and S. agalactiae. These tests are expensive small amount of CSF. High cost and availability only in
and their sensitivity is similar to that of Gram's smear. a few specialist laboratories are major limitations.
Owing to the occurrence of false-positive results, these CSF PCR is mainly useful for diagnosis of viral
tests are not recommended for routine diagnosis of infections of CNS (e.g. herpes simplex, enteroviruses,
meningitis. These tests are particularly useful in patients herpes zoster, etc.) and of tuberculous meningitis.
who have been partially treated and in whom Gram's CSF findings in different types of meningitis are
stain and culture are negative.
shown in Table 6.4.
Latex agglutination tests for cryptococcal antigens
are also available and have sensitivity of 90%. These tests 6. Special Investigations
have largely replaced India ink preparation for diagnosis (a) CSF protein electrophoresis: Protein electrophoresis
of cryptococcal meningitis. of normal CSF differs from normal serum in (i) presence
(e) Limulus lysate assay for endotoxin produced by of a prominent transthyretin band and (ii) an extra
gram-negative bacteria: Limulus amebocyte lysate assay transferrin band (called β2-transferrin or tau protein).
is a rapid, sensitive, and specific test for the presence of Protein electrophoresis of CSF is used:
Examination of Cerebrospinal Fluid 89

Table 6.4: Cerebrospinal fluid findings in different types of meningitis

Condition Appearance Leukocytes Proteins in Glucose in Additional


mg/dl mg/dl investigations

1. Normal Clear, <5/μl (mostly lymphocytes) 15-45 45-80 –


colorless
2. Acute pyogenic Turbid or Increased (>1000/μl); Increased; Decreased; Gram's stain;
meningitis purulent mostly neutrophils 50-1500 <40 culture; Latex
agglutination test
3. Tuberculous Clear or cloudy Increased (100-600/μl); Increased; Decreased; AFB stain; culture;
meningitis mostly lymphocytes or both 45-300 10-45 polymerase chain
lymphocytes and neutrophils reaction
4. Viral meningitis Clear or cloudy Increased (6-300/μl); Increased Normal Polymerase chain
lymphocytes reaction

Fig. 6.11: An example of positive tau protein: Lane 1: Normal


CSF; Lane 2: Serum; Lane 3: Nasal fluid. Lane 3 (nasal fluid)
shows two bands in the same position as normal CSF
confirming CSF leakage from nose

identification of such fluid as CSF is essential.


Fig. 6.10: Oligoclonal bands in CSF in gamma region Recommended test for this purpose is protein
(arrows) in a case of multiple sclerosis
electrophoresis with immunofixation for transferrin.
Protein electrophoresis of CSF shows an extra
• For identification of oligoclonal bands, and
transferrin band (called as 'tau' protein), which is
• To determine whether fluid submitted for
examination is CSF. absent in other body fluids and secretions. Tau protein
1. Oligoclonal bands: Agarose gel electrophoresis of is an enzymatically modified transferrin that moves
concentrated CSF is used for detection of oligoclonal just behind the unaltered transferrin in β region.
bands. These are two or more discrete bands in the Identification of two isoform bands of transferrin on
gamma region. Presence of oligoclonal bands on CSF protein electrophoresis is a highly sensitive and
protein electrophoresis that are absent on specific test for identification of fluid as CSF (Fig.
concurrently run serum protein electrophoresis is 6.11). Other body fluids or secretions do not show
indicative of intrathecal syntheis of immunoglobulins the second isoform band.
(Fig. 6.10). Oligoclonal bands have been identified in
majority of patients (90%) with multiple sclerosis; (b) Measurement of albumin and immunoglobulin G
however, they are not specific for this disorder. They (IgG): Comparison of IgG and albumin CSF/plasma ratio
are also seen in subacute sclerosing panencephalitis, can be helpful for diagnosis of multiple sclerosis (high
viral CNS infections, neurosyphilis, and Guillain- ratio due to intrathecal synthesis of IgG).
Barré syndrome.
2. CSF leakage: Occasionally, clear fluid leaking through REFERENCE RANGES
the nose or ear after trauma or surgery is submitted Reference ranges are given at the beginning of this
for examination to determine whether it is CSF. Due chapter under "Composition of normal cerebrospinal
to the risk of recurrent meningitis, accurate fluid in adults".
90 Essentials of Clinical Pathology

CRITICAL VALUES • Detection of blast cells or malignant cells


• Positive polymerase chain reaction for herpes simplex
• Cells: >10/cmm
• Glucose: <45 mg/dl BIBLIOGRAPHY
• Proteins: >45 mg/dl
• Detection of pathogens on Gram stain, latex aggluti- 1. Ellenby MS et al. Lumbar puncture. N Engl J Med 2006;
355:e12. Downloaded from www.nejm.org on January 14,
nation tests, Ziehl-Neelsen stain, or India ink
2007.
preparation 3. Seehusen DA, Reeves MM, Fomin DA. Cerebrospinal
• Positive culture fluid analysis. Am Fam Physician 2003;68:1103-8.
2. Riordan FAI, Cant AJ. When to do a lumbar puncture.
Arch Dis Child 2002;87:235-7.
7
Examination of Pleural and
Peritoneal Fluids

PLEURAL FLUID Causes of Pleural Effusion


Pleural cavity is a potential space between chest wall and There are various causes of pleural effusion (Table 7.1).
lung lined by a layer of flattened epithelium called as Common causes include congestive cardiac failure,
mesothelium. It contains a small amount of fluid (<10 bacterial pneumonia, tuberculosis, cirrhosis with ascites,
ml in each pleural cavity) which is an ultra-filtrate of and cancer. Cause of pleural effusion may be evident
plasma and which lubricates the two opposing parietal clinically such as congestive cardiac failure (raised
and visceral pleural layers. Pleural fluid is being formed jugular venous pressure, edema of feet), cirrhosis of liver
continuously depending on hydrostatic pressure, plasma (ascites, other signs of portal hypertension), lymphoma
colloid oncotic pressure, and permeability of capillaries or carcinoma (enlargement of lymph nodes, liver or
in parietal pleura. Resorption of pleural fluid occurs spleen), and pulmonary embolism (deep vein thrombosis
through lymphatics and venules in visceral pleura. of lower limbs, hemoptysis, pleuritic type of chest pain,
Composition of pleural fluid is similar to that of plasma, and dyspnea out of proportion to amount of fluid
except that it has lower protein level (< 1.5 gm/dl). accumulated). Accumulated pleural fluid may be either
Normal pleural fluid is clear and straw or pale yellow in a transudate or an exudate. Transudates result from non-
color. inflammatory conditions and are due to increase in
Accumulation of excess fluid in pleural cavity is called capillary hydrostatic pressure (e.g. congestive cardiac
as pleural effusion (Fig. 7.1). Presence of pleural effusion failure, portal hypertension) or low oncotic pressure of
is usually confirmed by lateral decubitus chest X-ray or plasma (e.g. hypoproteinemia). Exudates result from
ultrasonography. Radiography can also provide clues inflammatory conditions and are due to increased
about the cause of effusion e.g. bilateral effusion with capillary permeability (e.g. infections) or obstruction of
enlarged heart suggests congestive cardiac failure, pleural lymphatics (e.g. malignancy). Further testing of
radiological signs of pneumonia suggest parapneumonic transudates is not required, while it is necessary to
effusion. analyze exudates further. Comparative features of
transudates and exudates are presented in Table 7.2.

Collection of Pleural Fluid

Procedure for collection of pleural fluid by inserting a


needle in the chest wall is called as thoracentesis. It may
be either diagnostic or therapeutic. Diagnostic thora-
centesis is carried out to determine the cause of pleural
effusion. Conditions that can be definitely diagnosed are
empyema, malignancy, tuberculous effusion, fungal
effusion, esophageal rupture, chylothorax, hemothorax,
rheumatoid pleurisy, and lupus pleurisy. It is
recommended to perform diagnostic thoracentesis when
Fig. 7.1: Accumulation of fluid in pleural cavity on pleural effusion is clinically significant (i.e. >10 mm thick
X-ray chest on lateral decubitus chest X-ray or ultrasonography) and
92 Essentials of Clinical Pathology

Table 7.1: Causes of pleural effusion


Transudates Exudates

1. Congestive cardiac failure 1. Infections


2. Cirrhosis of liver • Parapneumonic effusion in bacterial pneumonia
3. Hypoproteinemia • Tuberculosis
4. Pulmonary embolism 2. Pulmonary infarction
3. Collagen disorders
• Rheumatoid arthritis
• Systemic lupus erythematosus
4. Acute pancreatitis
5. Trauma to chest wall
6. Malignancy
• Secondary deposits from bronchogenic carcinoma, carcinoma of
breast, etc.
• Lymphoma
• Mesothelioma

Table 7.2: Differences between transudate and exudate in pleural effusion

Parameters Transudates Exudates

1. Cause Non-inflammatory Inflammatory


2. Unilateral or bilateral Usually bilateral Usually unilateral
3. Mechanism Raised hydrostatic pressure or Increased capillary permeability or
reduced oncotic pressure in lymphatic obstruction
capillaries
4. Appearance Clear or straw-colored, does Turbid or cloudy, often clots
not clot
5. Specific gravity <1.016 >1.016
6. Proteins <3 gm/dl >3 gm/dl
7. Pleral fluid protein/serum protein ratio <0.5 >0.5
8. Lactate dehydrogenase (LDH) <200 >200
9. Pleural fluid LDH/serum LDH ratio <0.6 >0.6
10. Glucose Equivalent to plasma May be low
11. Cells Few lymphocytes and Many inflammatory cells
mesothelial cells
12. Organisms Nil May be present
13. Pleural fluid pH 7.45-7.55 <7.30

Note: No single criterion is adequate for differentiation. Test combinations are more sensitive and accurate

if cause is not obvious. Therapeutic thoracentesis is include introduction of infection, pneumothorax,


performed either to relieve respiratory insufficiency due hemothorax, air embolism, injury to the spleen or liver,
to massive pleural effusion or to introduce antineoplastic and pulmonary edema if large amount of pleural fluid
drugs in pleural space following withdrawal of pleural is rapidly removed.
fluid. For diagnostic studies, pleural fluid is collected in
heparinised tubes to prevent clotting. For cell counts,
A qualified and experienced physician performs
sample is collected in EDTA anticoagulant. For micro-
thoracentesis. It is contraindicated in non-cooperative
biologic studies, sterile sample tubes are necessary; for
patients, coagulation disorders, respiratory insufficiency, culture, aliquots of sample are best inoculated in blood
infection at the site of chest wall puncture, mechanical culture bottles at the bedside. In suspected malignancy,
ventilation, a small volume of fluid, unstable angina, and maximum amount of fluid should be submitted to
cardiac arrhythmias. Complications of the procedure enhance the cellular yield.
Examination of Pleural and Peritoneal Fluids 93

Examination of Pleural Fluid • Pleural fluid LDH above 2/3rds of upper limit of
normal for serum LDH.
This consists of:
Exudates have at least one of the above criteria.
• Appearance
Presence of all the three criteria best differentiates an
• Chemical examination exudate from a transudate. Transudates have none of
• Cell counts these criteria. If the fluid is an exudate, further
• Microbiological examination investigations are indicated like cell counts, estimation
• Cytological examination. of glucose, cytological examination, test for tuberculosis,
1. Appearance: Transudates are clear, pale yellow, or Gram smear, and culture. In case of transudates,
straw-colored and do not clot on standing. Exudates are diagnostic considerations include congestive cardiac
opaque or turbid due to increased proteins, leukocytes, failure, cirrhosis, and pulmonary embolism (Fig. 7.2).
or presence of malignant cells, and often clot on standing Low glucose levels are found in empyema,
due to high fibrinogen content. Frank pus with putrid rheumatoid pleurisy, tuberculous pleurisy, and
odor indicates empyema. malignant effusion.
Straw-colored transudative fluid occurs in congestive There is a direct correlation between lactate dehydro-
cardiac failure, pulmonary embolism, and cirrhosis of genase levels in pleural fluid with degree of pleural
liver. inflammation.
Thick exudative fluid occurs in pneumonia and 3. Cell counts: Total leukocyte count should routinely
cancer. be obtained on all fluids. In transudates, leukocytes are
Bloody or hemorrhagic fluid indicates traumatic tap, <1000/ml and are mostly small, mature lymphocytes.
pulmonary infarction, or malignancy. Hemorrhage in Predominance of neutrophils (>50%) indicates an
pleural space is referred to as hemothorax. A traumatic acute process (e.g. parapneumonic effusion, pulmonary
tap progressively becomes less bloodstained during infarct), while predominance of lymphocytes indicates
removal of pleural fluid. a chronic process (e.g. tuberculosis, rheumatoid pleurisy,
Accumulation of a milky or chylous pleural fluid is malignancy).
referred to as chylothorax. True chylous effusion results Very high leukocyte count (>50,000/ml) with
from obstruction of lymphatic duct (due to injury, predominance of neutrophils suggests parapneumonic
effusion (usually empyema). Presence of many small,
carcinoma, or lymphoma). A characteristic feature of
mature lymphocytes with only a few or no mesothelial
chylous effusion is triglyceride level >110 mg/dl.
cells is suggestive of tuberculosis.
Microscopy of such fluid shows lymphocytosis and fat
Blood-tinged pleural fluid is not diagnostic of any
droplets that stain with Sudan III. Following
condition and can occur in a transudate or an exudate.
centrifugation of a chylous fluid, a layer of creamy
Grossly hemorrhagic pleural fluids have red cells above
chylomicrons forms at the top. A pseudochylous effusion 100,000/ml and are seen in pulmonary infarction,
results from long-standing chronic pleural effusion malignancy, or traumatic tap. Traumatic bloody effusion
through breakdown of cellular lipids (tuberculous or is suggested by gradual clearing with aspiration, clotting
rheumatoid effusion). It has a bright, shiny appearance, of fluid after some time, and lack of hemosiderin-laden
and microscopic examination shows mixed cellularity macrophages.
and cholesterol crystals. Triglyceride level is ≤50 mg/dl
4. Microbiological examination: Gram’s smear and
and chylomicrons are absent.
culture should be carried out on exudates, especially on
A foul smelling pleural fluid may indicate a possible
purulent, bloodstained, or cloudy samples. The
anerobic infection, while urinous (ammoniacal) odor
likelihood of isolation of organisms is increased if pleural
indicates possible urinothorax.
fluid is inoculated in blood culture bottles at the bedside.
In mesothelioma, pleural fluid is viscous. Ziehl-Neelsen stained smear is positive in <20% of
2. Chemical examination: If old criteria are used for tuberculous pleural effusions and culture in <40% of
differentiation of exudates and transudates (like protein cases. If tuberculosis is suspected and culture is negative,
level, specific gravity, cell count, and clots in sample), polymerase chain reaction for mycobacterial DNA or
significant cases are misclassified. Therefore, in pleural increased level of adenosine deaminase (released in
fluid, differentiation of exudates from transudates is pleural fluid from activated lymphocytes) can establish
based on following criteria (known as Light’s criteria): the diagnosis.
• Pleural fluid protein/serum protein ratio > 0.5 5. Cytological examination: For cytological examination,
• Pleural fluid LDH/serum LDH ratio > 0.6 pleural fluid is centrifuged, smears are prepared from
94 Essentials of Clinical Pathology

Fig. 7.2: Evaluation of pleural effusion

the sediment, and are stained with Papanicolaou stain. Pleural fluid triglyceride ≥ 110 mg/dl indicates chylous
In older age, metastatic malignancy is responsible for effusion.
majority of pleural effusions. The common malignancies
are bronchogenic carcinoma, carcinoma of breast, and Box 7.1: Special studies on pleural fluid to determine the
cause of pleural effusion
lymphoma. Pleural effusion results from lymphatic
obstruction by tumor cells. Typically, effusion is blood • Tuberculous pleural effusion: Adenosine deaminase,
tinged or hemorrhagic. Examination of three separately gamma interferon, polymerase chain reaction
obtained pleural fluid samples increases sensitivity of • Rheumatoid effusion: Rheumatoid factor
detection of cancer cells to 80%.
• Lupus pleuritis: Antinuclear antibodies
6. Other tests: For diagnosis of pulmonary embolism,
• Pancreatic disease, esophageal disease: Amylase
D-dimer test (a breakdown product of fibrin) should be
• Malignancy: Carcinoembryonic antigen
carried out on peripheral blood sample. Flow cytometric
• Chylothorax: Triglycerides
analysis of pleural fluid sample should be done if
lymphoma is suspected. In parapneumonic effusion, if
pleural fluid pH is <7.20, drainage is indicated; if pH is Pleural fluid findings in main causes of pleural
>7.30, complete resolution will occur with medical effusion are shown in Table 7.3. Evaluation of pleural
treatment. In malignant pleural effusion, low pH effusion is shown in Figure 7.2. Special studies on pleural
indicates poor prognosis and poor response to fluid are listed in Box 7.1.
tetracycline pleurodesis. A pleural fluid rheumatoid
factor ≥ 1:320 is suggestive of rheumatoid pleurisy. PLEURAL BIOPSY
Presence of LE cells in pleural fluid makes diagnosis of Percutaneous needle biopsy of the parietal pleura is
lupus nephritis virtually certain. Elevated pleural fluid indicated in patients with exudative pleural effusion if
amylase occur in pancreatitis and esophageal rupture. examination of pleural fluid does not yield a specific
Examination of Pleural and Peritoneal Fluids 95

Table 7.3: Pleural fluid findings in main causes of pleural effusion

Causes Appearance Type of fluid Cells Special studies

1. Tuberculosis Straw-colored Exudate 5000-10000/μl; mostly Ziehl-Neelsen smear;


lymphocytes; <5% culture; polymerase
mesothelial cells chain reaction
2. Parapneumonic effusion* Turbid Exudate 5000-40000/μl; mostly Gram stain; culture
neutrophils
3. Empyema** Purulent Exudate >25000/cmm; mostly Gram stain; culture; low
neutrophils pH; low glucose
4. Malignancy*** Bloody Exudate Lymphocytes, Cytology, pleural biopsy
malignant cells
5. Congestive cardiac failure Clear Transudate Few mesothelial cells –

*Parapneumonic effusion: an exudative pleural effusion that occurs in bacterial pneumonia due to inflammation of pleura
adjacent to the affected lobe; **Empyema: Presence of purulent exudate in pleural cavity; it can result from parapneumonic
effusion, rupture of lung abscess in pleural space, injury, or rupture of subdiaphragmatic or hepatic abscess in pleural space;
*** Common causes of malignant pleural effusion are carcinoma of lung, carcinoma of breast, and lymphoma

diagnosis. It is done especially if tuberculosis or procedure can be combined with aspiration of pleural
malignancy is suspected. For better results, thoracoscopy fluid, pleural biopsy, and pleurodesis. Thorascopy is
(direct visualization of pleural surface by introducing a better than blind pleural biopsy for diagnosis of
thoracoscope through the chest wall into the pleural malignancy.
cavity previously filled with air) can be helpful in Thoracoscopy is contraindicated if pleural space is
localizing the site of biopsy. For diagnosis of tuberculosis, obliterated and in the presence of coagulopathy and
pleural biopsy is more sensitive than pleural fluid severe cardiac disease.
examination and culture. In malignancy, improved Complications include subcutaneous emphysema,
techniques in pleural fluid cytology have increased the empyema, spread of malignant cells along the access
diagnostic yield to 80%; combination of pleural fluid track, and hemorrhage. Reported mortality rate is <0.1%.
cytology and pleural biopsy can increase the detection
PERITONEAL FLUID
rate further.
For pleural biopsy, special needles like Abrams, Cope Peritoneal cavity is a potential space in the abdomen lined
and tru-cut are available. Needle is inserted with its by mesothelial cells and normally containing about 30-
cutting chamber closed into the pleural effusion. It is 50ml of serous fluid. The fluid is an ultrafiltrate of plasma
recommended to obtain at least three biopsy samples and its formation is dependent upon hydrostatic pressure,
(from 3, 6, and 9 ‘O’ clock positions of cutting chamber) plasma oncotic pressure, and capillary permeability.
for histology and culture. Pathological accumulation of fluid in peritoneal cavity is
Risks of hemothorax and pneumothorax are higher called as ascites, and the accumulated fluid is called as
ascitic fluid.
as compared to thoracentesis. Spread of malignant cells
can occur along the needle track. Causes of Ascites
Thoracoscopy: In thoracoscopy, an endoscope is Causes of ascites are listed in Table 7.4. Causes are
introduced into the pleural cavity for direct visualization classified based on whether the fluid is a transudate or
of pleural surfaces. It is indicated if pleural fluid analysis an exudate. Unlike pleural fluid, there are no well-
is non-diagnostic in a case of pleural effusion and to defined criteria for distinction between transudates and
localize the site of biopsy. exudates. Majority of patients with ascites have cirrhosis
Adequate pleural space is created by removing of liver; presence of ascites in a patient with cirrhosis is a
pleural fluid and replacing it with an equal amount of poor prognostic sign.
atmospheric air. Thoracoscope is inserted through a skin
incision via a trocar into the pleural cavity. Indications for Abdominal Paracentesis
It is especially helpful for diagnosis of metastatic Abdominal paracentesis refers to removal of ascitic fluid
cancer and mesothelioma. Appearance of pleura is also through puncture of the peritoneal cavity. Indications
diagnostic in tuberculosis and rheumatoid pleurisy. The for abdominal paracentesis are given in Table 7.5.
96 Essentials of Clinical Pathology

Table 7.4: Causes of ascites

Transudate (increased hydrostatic pressure or plasma Exudate (increased capillary permeability or


oncotic pressure) lymphatic obstruction)

1. Cirrhosis of liver 1. Bacterial peritonitis (primary or secondary)


2. Congestive cardiac failure 2. Tuberculosis
3. Hypoproteinemia 3. Malignancy (lymphoma, hepatoma, metastatic
carcinoma)
4. Abdominal injury
5. Biliary peritonitis (rupture of gallbladder)
6. Pancreatitis
7. Chylous ascites (obstruction of or injury to thoracic
duct)

Table 7.5: Indications for abdominal paracentesis


1. All patients with new-onset ascites
2. At admission in all patients with ascites for detection of
asymptomatic infection
3. All patients with ascites who develop clinical features
of bacterial infection, hepatic encephalopathy,
gastrointestinal hemorrhage, or impairment of renal
function.
4. Symptomatic ascites (therapeutic paracentesis)

Collection of Sample
Presence of ascites can usually be detected by clinical
examination; if clinical examination is not definitive,
ultrasound can be helpful. Ultrasonography can also be
useful for determining the cause of ascites.
A hollow needle is inserted through the abdominal Fig. 7.3: Sites for paracentesis (blue filled circles)
wall (usually left lower quadrant of abdomen below the
border of shifting dullness) into the peritoneal cavity Box 7.2: Tests commonly done on ascitic fluid
(Fig. 7.3) and fluid (20-50 ml) is removed under aseptic • White cell count: This is the most important test as it
precautions. For cytology, to maximize the yield of rapidly provides information about possible bacterial
malignant cells, 100 ml should be submitted. For cell infection. Neutrophil count >250/cmm is a strong
count sample is collected in EDTA-containing tube. For indication of bacterial infection, whereas lymphocytosis
microbiologic culture, sample is inoculated in blood indicates peritoneal tuberculosis or carcinomatosis.
culture bottles at bedside. • Albumin: Estimation of serum and ascitic fluid albumin
Complications of the procedure include hemorrhage, allows calculation of serum-ascites albumin gradient
perforation of viscus, and introduction of infection. (SAAG) that allows categorization of ascites into low and
high SAAG.
Evidence of fibrinolysis or of disseminated intra-
vascular coagulation in liver disease is a contraindication • Microbiological tests: Gram stain, Ziehl-Neelsen stain,
culture
for paracentesis.
• Cytological examination: For detection of malignant
Examination of Ascitic Fluid cells when peritoneum is involved by cancer.

Laboratory analysis of ascitic fluid helps in the 1. Appearance: Transudates are pale yellow or straw-
differential diagnosis of ascites. A variety of tests can be colored and clear, whereas exudates are opaque or turbid.
carried out; however, the tests should be decided in an Turbid fluid results from leucocytes, malignant cells, or
individual patient according to the clinical presentation. proteins. Bloody or hemorrhagic fluid indicates
The commonly performed tests include estimation of traumatic tap, recent surgery, abdominal trauma, or
total proteins and albumin, cell count, cytological malignancy. A traumatic tap shows gradual clearing of
examination, and bacterial culture (Box 7.2). fluid during aspiration. Milky or chylous fluid results
Examination of Pleural and Peritoneal Fluids 97

Fig. 7.4: Classification of ascites into high and low albumin gradient. This system has replaced test for total protein concentration
in ascitic fluid for classification of ascites into transudate and exudate. Patients with high albumin gradient respond to sodium
restriction and diuretics, while patients with low albumin gradient require specific treatment

from obstruction of lymphatic duct due to inflammation <1.0 gm/dl) from secondary bacterial peritonitis
or malignancy (lymphoma, carcinomatosis), or from (total protein > 1.0 gm/dl).
abdominal injury. ii. Lactate dehydrogenase: Lactate dehydrogenase in
ascitic fluid is elevated in spontaneous bacterial
2. Chemical examination: peritonitis (i.e. there is no obvious source of
i. Proteins: Traditionally, fluid is called as a transudate infection), secondary bacterial peritonitis (i.e.
if protein content is low, and an exudate if its protein identifiable source of infection is present), and in
content is high. However, this criterion alone is not peritoneal carcinomatosis.
always sufficient. In ascitic fluid, distinction Ascitic fluid findings in various diseases are shown
between transudates and exudates cannot be in Table 7.6. Distinction between spontaneous and
secondary bacterial peritonitis is presented in Table
reliably made by estimation of proteins. A better
7.7 and Figure 7.5.
indicator is albumin gradient (calculated as serum
iii. Amylase: Normally, amylase in ascitic fluid is
albumin minus ascitic fluid albumin done on the similar to serum amylase. If ascitic fluid amylase is
same day) (Fig 7.4). three times greater than serum amylase, ascites is
Total protein concentration in ascitic fluid can be most likely to be due to pancreatic disease such as
helpful in differentiating spontaneous (total protein acute pancreatitis.

Table 7.6: Ascitic fluid findings in various diseases

Cause Appearance Type of fluid Cells Special studies

1. Spontaneous bacterial peritonitis Cloudy or turbid Exudate Neutrophils Single organism isolated
≥250/μl on culture; Proteins
<1.0 gm/dl; Glucose normal
2. Secondary bacterial peritonitis Cloudy or turbid Exudate Neutrophils Multiple organisms isolated
≥1000/μl on culture
3. Cirrhosis of liver Clear, straw- Transudate Lymphocytes Albumin gradient ≥1.1 gm/dl
colored <500/μl
4. Tuberculous peritonitis Serosanguineous Exudate Lymphocytes Ziehl-Neelsen stain; culture;
>500/μl Low albumin gradient;
Total proteins ≥2.5 gm/dl
5. Malignancy Bloody Exudate Lymphocytes, Cytology
malignant cells
98 Essentials of Clinical Pathology

Table 7.7: Distinction between spontaneous and secondary bacterial peritonitis


Parameter Spontaneous bacterial peritonitis Secondary bacterial peritonitis

1. Obvious source of infection Absent Present, e.g. perforation of viscus, abscess


2. Total ascitic fluid proteins <1.0 gm/dl ≥1.0 gm/dl
3. Severity Less severe More severe
4. Culture Single organism Multiple organisms
5. Treatment Rapid response to antibiotics Requires surgical treatment

Fig 7.5: Differentiation of spontaneous from secondary bacterial peritonitis

iv. Bilirubin: Ascitic fluid bilirubin greater than 6.0 mg/ 50% of cases. Laparoscopic biopsy is more helpful in
dl and ascitic fluid bilirubin/serum bilirubin ratio diagnosis of tuberculous peritonitis.
greater than 1.0 indicate perforation of biliary tract 5. Cytological examination: Cytological examination of
(biliary peritonitis). Ascitic fluid is bile-stained. peritoneal fluid can detect 40-65% cases of malignant
3. Cell count: Cell count is usually done to distinguish ascites.
cirrhotic ascites from spontaneous bacterial
peritonitis. In ascitic fluid, total leukocyte count > BIBLIOGRAPHY
500/ml and absolute neutrophil count >250/ml 1. Light RW. Pleural effusion. N Engl J Med 2002;346:
constitute the presumptive evidence of spontaneous 1971-77.
bacterial peritonitis. 2. Runyon BA. Care of patients with ascites. N Engl J Med
4. Microbiological examination: Gram smear is positive 1994;330:337-42.
in 25% cases of spontaneous bacterial peritonitis. If 3. Tarn AC and Lapworth R. Biochemical analysis of
ascitic fluid is inoculated in blood-culture bottles pleural fluid; What should we measure? Ann Clin
at bedside, sensitivity of isolation rises to 85% (as Biochem 2001;38:311-22.
compared to conventional method of inoculation in 4. Thomsen TW, DeLaPena J, Setnik GS. Thoracentesis.
broth and agar plates in laboratory). In spontaneous N Engl J Med 2006;355:e16. Downloaded from
bacterial peritonitis, a single organism is isolated, www.nejm. org on January 14, 2007.
while secondary bacterial peritonitis is polymicrobial. 5. Thomsen TW, Shaffer RW, White B, Setnik GS.
In case of tuberculosis, Ziehl-Neelsen stain has Paracentesis. N Engl J Med 2006;355: e21. Downloaded
sensitivity of 25-30%, while culture is positive in about from www.nejm.org on January 14, 2007.
8

Examination of Sputum

Sputum examination refers to the examination of the Induction of Sputum


material coughed out from the lungs, bronchi, trachea,
If the patient is unable to bring out the sputum
and larynx. Normally, sputum is composed predomi-
spontaneously, inhaling aerosol of 15% sodium chloride
nantly of mucus and also certain cellular and non-cellular
and 20% propylene glycol for 20 minutes can induce
components of host origin. During expectoration, sputum
gets contaminated with cells and normal bacterial flora expectoration. Sputum can also be induced by inhaling
from the mouth and pharynx. nebulized hypertonic saline or distilled water in
Examination of sputum is mainly carried out for: association with chest physiotherapy.
• Identification of the causative organism in a For microbiologic studies, sample should be sent to
suspected infection of the lower respiratory tract, e.g. the laboratory immediately. If sputum is allowed to
– Pneumonia especially if severe or in an immuno- stand, rapid proliferation of contaminating bacterial flora
compromised host. from oral cavity and throat will occur leading to incorrect
– Suspected tuberculosis results. In addition, pathogenic organisms, especially
– Suspected fungal infection Haemophilus influenzae, do not survive in collected
– Pneumocystic carinii pneumonia in HIV-positive samples for long. Sample for bacterial culture should not
patients
be refrigerated.
– Infective exacerbation of a chronic disease like
If sample is to be transported to a distant laboratory
bronchiectasis
• Cytologic examination for malignant cells, investi- for mycobacterial culture, sputum should be collected
gation of asbestosis, investigation of viral infections in 25 ml of following solution:
(viral inclusions in cytomegalovirus and herpes • N-acetylpyridinium chloride 5 gm
simplex infections) and fungal infections. • Sodium chloride 10 gm
• Distilled water upto 1000 ml
COLLECTION OF SPUTUM
1. Sputum sample is preferably collected in the morning APPEARANCE OF SPUTUM
(since secretions accumulate overnight), soon after Appearance of sputum is often suggestive of the
awakening and before taking any mouthwash or underlying pathologic process as follows:
food.
• Bloody: Hemoptysis (pulmonary tuberculosis, lung
2. Sample is collected in a clean, dry, wide-mouthed
abscess, bronchiectasis, bronchogenic carcinoma,
container with a securely fitting screw cap. The
mitral stenosis, pulmonary infarction)
container should be leak proof and break-resistant to
• Rusty: Pneumococcal lobar pneumonia
prevent aerosol formation and desiccation, and
should have the capacity of about 25 ml. • Bloody and gelatinous (red current jelly): Klebsiella
3. Patient takes a deep breath 2-3 times filling his/her pneumonia
lungs, coughs deeply, and spits into the container. • Green: Pseudomonas infection
About 2-5 ml of sputum is collected. Sample • Purulent and separating into 3 layers on standing:
consisting only of saliva (foamy, clear, and watery Bronchiectasis, lung abscess
appearance) is not acceptable; in such a case, another • Pink, frothy (air bubbles): Pulmonary edema
sample should be obtained. The container is capped • Copious amounts of purulent sputum: Lung abscess,
securely and labeled. bronchiectasis, bronchopleural fistula
100 Essentials of Clinical Pathology

MICROBIOLOGICAL EXAMINATION Gram stained smear of sputum should be interpreted


carefully because of the presence of various contami-
Sputum sample is often contaminated with normal flora nating gram-positive and gram-negative organisms
of the oral cavity and pharynx (Box 8.1). originating from mouth and throat (normal bacterial
flora).
Box 8.1: Normal flora of the oral cavity and pharynx Morphological appearance on Gram stained smear
is suggestive of a particular organism as follows:
Gram-positive: Staphylococci (S. aureus, S. epidermidis), • Gram-positive diplococci with surrounding clear
streptococci (S. viridans, S. pneumoniae), Diptheroids,
space (capsule): S. pneumoniae (Fig. 8.2).
enterococci, micrococci, lactobacilli, Yeasts (Candida spp.).
• Gram-positive cocci in grape-like clusters: S. aureus.
Gram-negative: Neisseria spp; Haemophilus spp; fuso- • Gram-positive yeast cells with budding and pseudo-
bacteria, coliforms, Moraxella catarrhalis.
hyphae: Candida.
• Gram-negative diplococci, both intra- and extra-
cellular: Moraxella catarrhalis.
Gram Staining
• Gram-negative coccobacilli: H. influenzae.
Pathogenic organisms found in sputum include— • Large granules with center gram-negative and
1. Gram-positive: Streptococcus pneumoniae, Streptococcus periphery gram-positive: Actinomyces.
pyogenes, Staphylococcus aureus.
2. Gram-negative: Haemophilus influenzae, Klebsiella Bacteriological Culture
pneumoniae, Pseudomonas aeruginosa, Yersinia pestis, A floccule of purulent portion of sputum is inoculated
Moraxella catarrhalis. on culture media for definitive identification of
For bacteriologic examination, sputum sample should organisms. Sputum sample is considered as unsatis-
be processed in the laboratory within 1 hour of collection. factory for culture if it contains >25 squamous cells/low
The sample is transferred to a sterile Petri dish and its power field. Ideal sputum sample for culture contains
appearance is noted. A thin smear is made on a glass alveolar macrophages, numerous neutrophils (>5/high
slide from the purulent portion of the sputum with a power field), bronchial epithelial cells, and few squamous
clean stick, air-dried, fixed, and stained with Gram’s cells (<10/high power field). To reduce the amount of
stain. Purely mucoid, watery, frothy, or white samples contaminating normal bacterial flora in the inoculum,
usually show squamous epithelial cells covered with saliva is washed away from sputum with sterile normal
masses of bacteria; this indicates that the sample consists saline. The washed sputum is inoculated on (i) blood agar
mainly of secretions from the mouth and the throat. Such plate and (ii) chocolate agar (heated blood agar). The
samples are not suitable for bacteriological examination blood agar plate is incubated aerobically and chocolate
(Fig. 8.1). If polymorphonuclear neutrophils are less than agar plate is incubated in an atmosphere of extra carbon
10 per epithelial cell, culture is not carried out. dioxide. Inoculated plates are inspected for growth after

Fig. 8.1: Unacceptable sputum sample: Sputum sample Fig. 8.2: Gram stained smear of sputum showing
shows many squamous cells covered with masses of bacteria gram-positive diplococci (Streptococcus pneumoniae)
Examination of Sputum 101

incubation for 18 hours; if growth is not satisfactory, Ziehl-Neelsen stain of sputum smear: This simple,
incubation for further 24 hours is indicated. Antibiotic inexpensive, and rapid technique is mainly useful for:
sensitivity test is carried out only if amount of growth is • Diagnosis of infectious cases of pulmonary tubercu-
significant. losis. (Sputum smear-positive cases are a major source
of spread of infection).
EXAMINATION OF SPUTUM FOR • Assessment of response to anti-tuberculous treat-
MYCOBACTERIUM TUBERCULOSIS ment.
• Determining cure or treatment failure.
Tuberculosis is one of the major public health problems Ziehl-Neelsen-stained sputum smear is positive if at
in India. Early diagnosis of pulmonary tuberculosis will least 5000-10000 tubercle bacilli/ml are present in the
lead to early institution of therapy enabling cure, and sputum. Sensitivity of the technique is reported to be 60-
also prevention of spread of disease to others. Number 80%. Chances of detection of tubercle bacilli are increased
of cases of tuberculosis has increased in recent times, with if multiple sputum samples are examined or if bleach
World Health Organization calling it a global emergency. concentration technique is used. In bleach concentration
Multidrug-resistant tuberculosis is also emerging on a technique, a solution of bleach (concentrated sodium
large scale. hypochlorite) is added to the sputum sample, which
Mycobacterium tuberculosis complex comprises of M. leads to the liquefaction of mucus and killing of
tuberculosis, M. bovis, and M. africanum. These tubercle mycobacteria. After centrifugation (or overnight
bacilli are the aetiologic agents of human tuberculosis. sedimentation), smears are prepared from the sediment,
Other mycobacteria are called as non-tuberculous. stained, and examined.
There are two main approaches for diagnosis of With Ziehl-Neelsen staining, mycobacteria appear as
tuberculosis: bright red straight or slightly curved beaded rods (2-4 μ
• Direct tests: This consists of detection of M. in length and 0.2-0.5 μ in width) against a blue or green
tuberculosis or its components background (Fig. 8.3). Mycobacteria are both acid- and
• Indirect tests: This involves detection of humoral or alcohol-fast and are termed acid-fast bacilli (AFB). If acid-
cellular immune response to tuberculous infection. fast bacilli are seen, their number should be reported. At
These tests have not been considered in this chapter. least 100 fields are examined before reporting the smear
Direct tests for diagnosis of tuberculosis on sputum as negative.
A negative smear does not rule out the diagnosis of
sample are as follows:
tuberculosis since organisms may be few in number,
• Examination of sputum smear
sputum sample may not have been collected satisfac-
1. Ziehl-Neelsen technique torily, or smear may be of poor quality.
2. Fluorescence microscopy
• Culture on conventional media
• Commercial automated culture systems
• Molecular methods.

Examination of Sputum Smear

For detection of M. tuberculosis, at least three sputum


samples collected on three separate occasions (including
at least one early morning specimen) need to be
examined. A thin smear is prepared on a clean glass slide
from blood-tinged, opaque, grayish, or yellowish portion.
Children often swallow sputum and may not be able to
cough it up; in them a sample of fasting gastric juice can
be aspirated and examined like sputum.
The smear is stained with Ziehl-Neelsen stain and
examined under the ordinary light microscope. If
fluorescent microscope is available, smear can be
examined after staining it with a fluorochrome Fig. 8.3: Sputum smear stained with Ziehl-Neelsen stain
(auramine-rhodamine or auramine O). showing acid-fast bacilli
102 Essentials of Clinical Pathology

sample is carried out to liquefy the organic debris so that


the decontaminating agent can reach the bacteria, and
kill the contaminating organisms. The usual agent
employed for this purpose is 4% sodium hydroxide.
Traditional culture media for isolation of M.
tuberculosis are:
• Solid media: Egg-based (Lowenstein-Jensen medium)
or agar-based (Middlebrook 7H10 or 7H11).
• Liquid media: Middlebrook 7H9, Middlebrook 7H12.
The most commonly used solid medium for culture
is Lowenstein-Jensen medium. Visible mycobacterial
growth may require up to 6 weeks. Further biochemical
tests are required for determination of species.

Commercial Automated Culture Systems


Fig. 8.4: Demonstration of mycobacteria in sputum by New rapid automated culture systems have been
fluorescence microscopy. Bacilli appear as yellow fluorescing introduced commercially which can give result within 2
rods with auramine O against dark background weeks (instead of 6 weeks with conventional media).
These methods, however, are expensive. One such
system is BACTEC 460TB system (Becton-Dickinson
Fluorescence microscopy: M. tuberculosis can also be stained Diagnostic Instruments Systems, Maryland, USA). This
with a fluorochrome (auramine-rhodamine or auramine is a well-established, sensitive, and rapid method for
O) and the preparation is examined under fluorescence detection of M. tuberculosis in clinical samples. This
microscope. Mycobacteria fluoresce bright yellow against
method uses a broth in which radiolabeled 14C-palmitate
a dark background (Fig. 8.4). This technique is rapid
has been incorporated. Mycobacteria metabolize 14C-
(since the smear is examined under low power) and is
palmitate to radiolabelled 14CO2, which is then detected
especially helpful if organisms are few in number. It is
by the instrument.
necessary to confirm a positive smear by Ziehl-Neelsen
stain since false-positive rate is high. Molecular Methods
Culture (Conventional) There are two approaches for molecular diagnosis of
The definitive diagnosis of tuberculosis is based on tuberculosis in sputum samples:
isolation of M. tuberculosis from culture of sputum • Direct detection of Mycobacterium tuberculosis in
sample. Culture is usually carried out for: sputum sample
• Drug susceptibility testing • Detection of Mycobacterium tuberculosis in isolates
• Precise species identification, if organism other than from culture by nucleic acid probes.
M. tuberculosis is suspected (for epidemiologic M. tuberculosis can be rapidly identified directly in
purpose). sputum samples by detecting DNA sequences specific
• Diagnosis in patients who have typical clinical and to it. A commonly used DNA target for this purpose is
radiological features of tuberculosis but are sputum IS 6110 that is observed only in M. tuberculosis complex.
smear-negative. Multiple copies of this sequence are present in the
Culture is more sensitive than sputum smear exami- genome of M. tuberculosis. This method can detect 10-
nation as it can detect 10-100 microorganisms per ml of 1000 organisms per ml of sputum. Other DNA and RNA
sputum sample. Its sensitivity for diagnosis of tuber- sequences specific for M. tuberculosis complex can also
culosis is 80-85% and specificity is 98%. However culture be targeted.
is expensive, around 6 weeks are required for result and Laboratory cross-contamination (due to aerosolized
even longer for drug susceptibility testing, and prior PCR product) is responsible for significant number of
decontamination of sputum is required to kill normal false-positive results. This test is also expensive. PCR
bacterial flora. amplifies DNA sequences of both live and dead bacilli.
Sputum samples are contaminated to a varying Therefore the test cannot be used to assess response to
degree with normal bacterial flora. These bacteria are therapy.
rapidly growing and digest the culture medium before PCR-based assays should be interpreted in the light
tubercle bacilli begin to grow. Therefore, before of clinical features, prevalence of tuberculosis in the
inoculation, digestion and decontamination of sputum population, and findings on Ziehl-Neelsen smear.
Examination of Sputum 103

EXAMINATION FOR OTHER ORGANISMS


Additional investigations given below are indicated
when infection by following organisms is suspected:
• Pneumocystis carinii: Bronchoalveolar lavage fluid
stained with silver stain (Fig. 8.5) and Giemsa stain
• Yersinia pestis (pneumonic plague): Giemsa smear
• Yeast-like organisms on Gram’s smear: Sabouraud
dextrose agar
• Histoplasmosis: Giemsa smear
• Aspergillus: Potassium hydroxide wet mount of
sputum
• Paragonimus: Saline wet mount of sputum for eggs.

CYTOLOGICAL EXAMINATION OF SPUTUM


Cytological examination of sputum is usually carried out
for investigation of bronchogenic carcinoma. Sometimes,
it may also be helpful in the identification of viral
inclusions (like those of cytomegalovirus and Herpes Fig. 8.6: Sputum examination showing malignant cells
simplex virus), fungi, protozoa, and asbestos bodies. of squamous type
For cytological examination, fresh early morning
sputum sample is preferred. For detection of lung cancer, fixative. If delay is anticipated, prefixation of sputum
it is recommended to collect sputum sample daily for with Saccomano’s fixative is recommended. This consists
five consecutive days. This is because multiple sputum of collection of sputum in a mixture of 50% ethyl alcohol
samples increase the chances of detection of malignant and 2% carbowax.
cells (Fig. 8.6). In the laboratory, smears are made from blood-tinged
Sputum sample may be either spontaneously portion or from tissue fragments in sputum and stained
produced or artificially induced. If patient is unable to with Papanicolaou technique. Sputum sample is
produce sputum spontaneously by deep coughing, he is considered as adequate for cytological evaluation if
made to inhale a heated, aerosolized solution of 15% alveolar macrophages or bronchial epithelial cells are
sodium chloride and 20% propylene glycol for 20 seen in the smear.
minutes. This usually results in induction of a satisfactory The average sensitivity of sputum examination for
sputum sample. detection of malignant cells is about 65%. Sensitivity
Ideally, freshly collected sputum should be sent increases if:
immediately to the laboratory without addition of any • Increased numbers of sputum samples are examined
• Lesion is centrally located rather than at the periphery
of the lung
• Size of tumor is large
• Histologic type of carcinoma is of squamous nature
rather than adenocarcinoma or small cell carcinoma.
BIBLIOGRAPHY
1. American Thoracic Society. Diagnostic standards and
classification of tuberculosis in adults and children. Am
J Respir Crit Care Med 2000;161:1376-95.
2. Indian Council of Medical Research. What is new in
the diagnosis of tuberculosis? Part I: Techniques for
diagnosis of tuberculosis. ICMR Bulletin 2002;32;No 8
3. Laszlo A. Tuberculosis: Laboratory aspects of diagnosis.
CMAJ 1999;160:1275-9.
4. Soini H, Musser JM. Molecular diagnosis of myco-
bacteria. Clin Chem 2001;47:809-14.
5. Thunissen FBJM. Sputum examination for early
detection of lung cancer. J Clin Path 2003;56:805-10.
6. Watterson SA, Drobniewski FA. Modern laboratory
Fig. 8.5: Pneumocystis carinii cysts in bronchoalveolar diagnosis of mycobacterial infections. J Clin Path 2000;
lavage fluid (silver methenamine stain) 53:727-32.

tahir99 - UnitedVRG
vip.persianss.ir
9

Examination of Feces

Waste products excreted from the digestive tract are stool sample or 48- or 72-hour sample is collected.
composed of water (up to 75%), indigestible residue, Random diarrheal sample can be tested for occult
undigested food, food which is digested but not blood, fat, pH, white blood cells, culture, or micro-
absorbed, bile, epithelial cells, secretions from digestive scopy. A 48- or 72-hour sample is examined for
tract, inorganic material, and bacteria. Normal amount weight, fat content, carbohydrate, osmolality, or
of feces in an adult is 100-200 grams per day. chymotrypsin activity.
Examination of feces is helpful in the investigation • Evaluation of dysentery: Identification of causative
of diseases of the gastrointestinal tract as follows: organism is definitive in differentiating amebic from
• Detection of parasites: Stool examination is done for
bacillary dysentery.
detection of worms (adult worms, segments of
• Bacteriologic examination: Infection by bacteria such
tapeworms, larvae, ova), and protozoa (trophozoites
or cysts). as Salmonella, Shigella, Vibrio, Yersinia, or Clostridium
• Evaluation of chronic diarrhea: Chronic diarrhea difficile can be identified by stool culture. Bacterial
refers to passage of 3 or more loose or liquid stools toxins (such as those released by Clostridium botulinum
per day lasting for more than 4 weeks. Acute diarrhea or Clostridium difficile) can also be identified.
is defined as passage of 3 or more loose or liquid stools • Chemical examination: Chemical tests can be applied
per day for less than 4 weeks. In diarrhea, stool on feces to detect occult blood (in ulcerated lesions of
examination is an important part of laboratory work- gastrointestinal tract, especially occult carcinoma of
up. Causes of chronic and acute diarrhea are listed in colon), excess fat excretion (malabsorption syn-
Table 9.1 and Figure 9.1 respectively. Depending on drome), and presence or absence of urobilinogen
the nature of information sought, either a random (obstructive jaundice).

Table 9.1: Classification and causes of chronic diarrhea

1. Watery diarrhea
i. Osmotic
• Carbohydrate malabsorption
• Osmotic laxatives
ii. Secretory
• Bacterial toxins
• Bile acid malabsorption
• Laxative abuse
• Hormonal disorders: VIPoma, carcinoid syndrome, gastrinoma, hyperthyroidism
2. Inflammatory diarrhea
i. Invasive bacterial and parasitic infections
ii. Inflammatory bowel disease
iii. Pseudomembranous colitis
iv. Infectious diseases
v. Neoplasia
3. Fatty diarrhea
• Malabsorption syndromes

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Feces 105

Fig. 9.1: Classification of causes of acute diarrhea

Fig. 9.2: Preliminary evaluation of acute diarrhea. Examination of feces for white blood cells is helpful in
narrowing differential diagnosis in intestinal infections in acute diarrhea

• Differentiating infection by invasive bacteria (like rescence, or enzyme-linked immunosorbent assay


Salmonella or Shigella) from that due to toxin- (ELISA).
producing bacteria (like Escherichia coli or Vibrio
MICROSCOPIC EXAMINATION
cholerae): Increased numbers of polymorphonuclear
neutrophils (identified by methylene blue stain) are Microscopic examinations done on fecal sample are
shown in Figure 9.3.
seen in the former (Fig. 9.2).
• Identification of Rotavirus: Rotavirus is a common Collection of Specimen for Parasites
cause of diarrhea in infants and young children. It A random specimen of stool (at least 4 ml or 4 cm3) is
can be identified by examination of stool by electron collected in a clean, dry, container with a tightly fitting
microscopy, latex agglutination, immunofluo- lid (a tin box, plastic box, glass jar, or waxed cardboard

tahir99 - UnitedVRG
vip.persianss.ir
106 Essentials of Clinical Pathology

Fig. 9.3: Microscopic examinations carried out on fecal sample

box) and transported immediately to the laboratory (this


is because trophozoites of Entameba histolytica rapidly
degenerate and alter in morphology). About 20-40 grams
of formed stool or 5-6 tablespoons of watery stool should
be collected. Stool should not be contaminated with urine,
water, soil, or menstrual blood. Urine and water destroy
trophozoites; soil will introduce extraneous organisms
and also hinder proper examination. Parasites are best
detected in warm, freshly passed stools and therefore
stools should be examined as early as possible after Fig. 9.4: Consistency of feces
receipt in the laboratory (preferably within 1 hour of
collection). If delay in examination is anticipated, sample
Color/Appearance of Fecal Specimens
may be refrigerated. A fixative containing 10% formalin
(for preservation of eggs, larvae, and cysts) or polyvinyl • Brown: Normal
alcohol (for preservation of trophozoites and cysts, and • Black: Bleeding in upper gastrointestinal tract
for permanent staining) may be used if specimen is to be (proximal to cecum), Drugs (iron salts, bismuth salts,
transported to a distant laboratory. charcoal)
One negative report for ova and parasites does not • Red: Bleeeding in large intestine, undigested tomatoes
exclude the possibility of infection. Three separate or beets
samples, collected at 3-day intervals, have been • Clay-colored (gray-white): Biliary obstruction
recommended to detect all parasite infections. • Silvery: Carcinoma of ampulla of Vater
Patient should not be receiving oily laxatives, anti- • Watery: Certain strains of Escherichia coli, Rotavirus
diarrheal medications, bismuth, antibiotics like tetra- enteritis, cryptosporidiosis
cycline, or antacids for 7 days before stool examination. • Rice water: Cholera
Patient should not have undergone a barium swallow • Unformed with blood and mucus: Amebiasis,
examination. inflammatory bowel disease
In the laboratory, macroscopic examination is done • Unformed with blood, mucus, and pus: Bacillary
for consistency (watery, loose, soft or formed) (Fig. 9.4), dysentery
color, odor, and presence of blood, mucus, adult worms • Unformed, frothy, foul smelling, which float on
or segments of tapeworms. water: Steatorrhea.
Trophozoites are most likely to be found in loose
or watery stools or in stools containing blood and Preparation of Slides
mucus, while cysts are likely to be found in formed
stools. Trophozoites die soon after being passed and After receipt in the laboratory, saline and iodine wet
therefore such stools should be examined within 1 hour mounts of the sample are prepared (Fig. 9.5).
of passing. Examination of formed stools can be delayed A drop of normal saline is placed near one end of a
but should be completed on the same day. glass slide and a drop of Lugol iodine solution is placed

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Feces 107

Fig. 9.5: Saline and iodine wet mounts of fecal sample

near the other end. A small amount of feces (about the


size of a match-head) is mixed with a drop each of saline
Fig. 9.6: Formol-ethyl acetate concentration technique
and iodine using a wire loop, and a cover slip is placed
over each preparation separately. If the specimen
contains blood or mucus, that portion should be included • Floatation techniques: Ova and cysts float on surface.
for examination (trophozoites are more readily found in However, some ova and cysts do not float at the top
mucus). If the stools are liquid, select the portion from in this procedure. Examples: Saturated salt floatation
the surface for examination. technique and zinc sulphate concentration technique.
Saline wet mount is used for demonstration of eggs The most commonly used sedimentation method is
and larvae of helminths, and trophozoites and cysts of formol-ethyl acetate concentration method since: (i) it can
protozoa. It can also detect red cells and white cells. detect eggs and larvae of almost all helminths, and cysts
Iodine stains glycogen and nuclei of the cysts. The iodine of protozoa, (ii) it preserves their morphology well, (iii) it
wet mount is useful for identification of protozoal cysts. is rapid, and (iv) risk of infection to the laboratory worker
Trophozoites become non-motile in iodine mounts. A is minimal because pathogens are killed by formalin.
liquid, diarrheal stool can be examined directly without In this method, fecal suspension is prepared in 10%
adding saline. formalin (10 ml formalin + 1 gram feces). This suspension
is then passed through a gauze filter till 7 ml of filtered
Concentration Procedure material is obtained. To this, ethyl acetate (3 ml) is added
Concentration of fecal specimen is useful if very small and the mixture is centrifuged for 1 minute. Eggs, larvae,
numbers of parasites are present. However, in concen- and cysts sediment at the bottom of the centrifuge tube
trated specimens, amebic trophozoites can no longer be (Fig. 9.6). Above this deposit, there are layers of formalin,
detected since they are destroyed. If wet mount fecal debris, and ether. Fecal debris is loosened with an
examination is negative and there is clinical suspicion of applicator stick and the supernatant is poured off. One
parasitic infection, fecal concentration is indicated. It is drop of sediment is placed on one end of a glass slide
used for detection of ova, cysts, and larvae of parasites. and one drop is placed at the other end. One of the drops
Various concentration methods are available; the is stained with iodine, cover slips are placed, and the
choice depends on the nature of parasites to be identified preparation is examined under the microscope.
and the equipment/reagent available in a particular
laboratory. Concentration techniques are of two main Classification of
types: Intestinal Parasites of Humans
• Sedimentation techniques: Ova and cysts settle at the
bottom. However, excessive fecal debris may make Intestinal parasites of humans are classified into two
the detection of parasites difficult. Example: Formol- main kingdoms: protozoa and metazoa (helminths)
ethyl acetate sedimentation procedure. (Fig. 9.7).

tahir99 - UnitedVRG
vip.persianss.ir
108 Essentials of Clinical Pathology

Fig. 9.7: Classification of intestinal parasites of humans

PROTOZOA in submucosa, multiply, and cause disease (colitis).


Extraintestinal spread to liver and other sites can occur.
Entamoeba histolytica
Under unfavorable conditions, encystation of tropho-
E. histolytica is worldwide in distribution, and endemic zoites leads to the formation of cysts in the intestinal
in tropical and subtropical countries. It is transmitted by lumen. Cysts are discharged in feces and can survive in
fecal-oral route (ingestion of food or water contaminated moist environmental conditions for weeks to months and
by cysts of E. histolytica). propagate the life cycle by further fecal-oral spread.
Infection by E. histolytica may be asymptomatic or
may cause amebic dysentery or amebic liver abscess. Laboratory Diagnosis
Amebic trophozoites invade the large intestinal mucosa,
multiply in submucosa, spread laterally and produce 1. Identification of trophozoites and cysts on stool
flask-shaped ulcers. Symptoms include low-grade fever, examination: Demonstration of trophozoites of E.
diarrhea with blood and mucus, weight loss, and histolytica in fecal specimens is required for diagnosis
cramping abdominal pain. The cecum, ascending colon, of amebic dysentery. For diagnosis, at least three fresh
and rectosigmoid are commonly affected. Excessive stool samples should be examined to increase
granulation tissue may form in the intestine at the site of sensitivity. Trophozoites vary from 15 to 40 μ in
lesion (ameboma) to produce constriction, which may diameter. In saline wet mounts, trophozoites show
be mistaken clinically for a neoplasm. motility in one direction via pseudopodia, which form
In some cases, amebae penetrate the portal vessels rapidly. Cytoplasm shows outer transparent ecto-
and are transported to the liver where they form liver
plasm and inner finely granular endoplasm. Diag-
abscess. Amebic abscesses can also form in lungs or brain.
nostic feature of E. histolytica trophozoites is the
Life cycle: Infection is acquired by ingestion of food or presence of ingested red cells. Nucleus is visible in
water contaminated with infective (quadrinucleate) cysts the iodine preparation (Fig. 9.8). Fine granules of
of E. histolytica. Dissolution of cyst wall in the small peripheral nuclear chromatin are evenly distributed
intestine occurs with formation of trophozoites. Actively along the nuclear membrane. Karyosome is small and
motile trophozoites invade large intestinal mucosa, lodge centrally placed (Motility is lost in iodine mount).

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Feces 109

Fig. 9.8: Trophozoite (left) and uninucleate cyst (right) of E. histolytica. Nuclear chromatin is finely beaded and
evenly coats regular nuclear membrane. Karyosome is central. Chromatoid body is long with rounded ends

Cysts of E. histolytica are spherical and measure 10-


15 μ in diameter. Nuclei are 1, 2, 3, or 4 and are similar in
morphology to trophozoite nucleus (mature cyst contains
4 nuclei). Nuclear membrane is regular and thin with
finely granular peripheral chromatin. Karyosome is small
and central. Immature cysts may show chromatoid
bodies (aggregates of ribosomes) that are oblong
structures with rounded ends, and glycogen clumps
(Fig. 9.9).
Entameba dispar is a morphologically identical
organism; however, it is non-pathogenic. If red blood
cells are identified within trophozoites, then the species
is E. histolytica. However, there is no morphologic feature
to distinguish between the cysts of these two organisms.
Therefore, if only cysts are identified on stool exami-
nation, they are reported as “Cysts of E. histolytica/dispar”.
Asymptomatic infections with E. histolytica reported in
the past are now known to be due to E. dispar. Monoclonal
antibodies can distinguish between these two organisms.
Fig. 9.9: Uninucleate, binucleate, trinucleate, and
quadrinucleate cysts of Entamoeba histolytica
Entamoeba histolytica can be definitively identified in
saline wet mount if it shows definite directional motility
and contains ingested red cells.

It is also necessary to distinguish E. histolytica from


other non-pathogenic amebae found in stools like
Entamoeba coli, Entamoeba hartmanii, Endolimax nana, and
Iodameba butschlii.
Wet mount examination of stool has low sensitivity
(25-60%) and also false-positivity due to E. dispar.
For identification of trophozoites, stool smears can
be prepared and stained with trichrome stain (Fig. 9.10).
2. Other findings on stool examination: Plenty of red Fig. 9.10: Trophozoite of Entamoeba histolytica showing
cells and very few white cells are helpful in ingested red cells (Trichrome stain)

tahir99 - UnitedVRG
vip.persianss.ir
110 Essentials of Clinical Pathology

Table 9.2: Differences between amebic and bacillary dysentery

Parameter Amebic dysentery Bacillary dysentery

1. Cause Entamoeba histolytica Shigella (most common)


2. Onset Gradual Acute
3. Fever/vomiting Not significant Significant
4. Appearance of fecal sample Unformed with blood and mucus Unformed with blood, mucus, and pus
5. Microscopic examination of stool
• Red cells Clumps Discrete
• Pus cells Nil or few Numerous
• Macrophages Not seen Many, some with ingested red cells
• Charcot-Leyden crystals May be present Not seen
• Trophozoites of E. histolytica Present Not seen
• Bacteria Many, motile Few, nonmotile
6. Antigen test for E. histolytica Positive Negative
7. Stool culture Negative Positive for Shigella

differentiating amebic from bacillary dysentery. Giardia intestinalis (lamblia)


Charcot-Leyden crystals may be seen. Differences
G. intestinalis (lamblia), a pathogenic intestinal protozoan,
between amebic and bacillary dysentery are listed in has a worldwide distribution. It is transmitted by fecal-
Table 9.2. oral route, and is usually water-borne. Giardia is resistant
3. Detection of antigen of E. histolytica in stools: Direct to chlorine levels in tap water and is commonly found in
detection of antigen specific to E. histolytica is possible cold mountain streams. Giardiasis frequently occurs in
by commercially available tests based on enzyme persons who spend time camping outdoors or in woods
and is also called as “Backpacker’s diarrhea” or “beaver
immunoassay. These tests are specific and sensitive
fever”. It can cause asymptomatic infection, mild
(90%) and can differentiate E. histolytica from E. dispar.
diarrhea, or a severe disease with diarrhea, mal-
These tests are indicated if direct stool examination absorption, weight loss and steatorrhea.
is negative for organisms and intestinal amebiasis is There are two stages in the life cycle: cyst and
suspected clinically. trophozoite. After ingestion of cysts, excystation occurs
4. Detection of DNA specific to E. histolytica is possible due to action of gastric acid, and trophozoites are released
by polymerase chain reaction-based assays. which migrate to the duodenum and proximal jejunum
where they attach to the mucosa and replicate.
5. Serologic tests: Serologic tests, which detect anti-
bodies to E. histolytica, are performed to support the Laboratory Diagnosis
diagnosis of invasive amebiasis (i.e. colitis, liver 1. Demonstration of trophozoites or cysts: G. lamblia
abscess, or ameboma) when organisms cannot be trophozoites are found in fresh liquid stools,
demonstrated on stool examination. Various tests are particularly in flakes of mucus. They often occur in
available like latex agglutination test, indirect clusters. Cysts are more likely to be found in formed
hemagglutination test, enzyme immunoassay, and or loose stools.
counter immunoelectrophoresis. Enzyme immuno- Duodenal aspirates may be obtained if repeated
assay is the method of choice since it is most sensitive fecal examinations are negative for G. lamblia and
there is a strong clinical suspicion of giardiasis.
and specific. Serologic tests, however, remain positive
However, the test is invasive and is usually not
for many years after infection and thus cannot
necessary for diagnosis.
distinguish between recent and past infections. Trophozoites of G. lamblia are pear-shaped,
6. Endoscopic biopsy of ulcer in the intestine: This can 12-15 μ in diameter, have 4 pairs of flagellae, 2 large
demonstrate trophozoites of E. histolytica in 50% of and oval nuclei, 2 axonemes (axial filaments of
cases. Staining with periodic acid Schiff stain flagella), and 1 or 2 curved median bodies. Motility
facilitates identification of parasites. is likened to that of “falling leaf”.

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Feces 111

zoites develop into male and female gametes. Fertili-


zation of male and female gametes produces a zygote.
Oocyst is formed by encystation around the conjugating
gametes. Sporozoites form within oocysts by sexual
reproduction or sporogony. Oocysts are excreted in feces
and contaminate food or water.

Laboratory Diagnosis
1. Examination of stools for demonstration of oocysts
of I. belli, C. parvum, or C. cayetanensis:
Isospora belli: Oocysts of I. belli can usually be found
in direct wet mounts of feces (unstained). Formalin-
ether concentration technique may sometimes be
necessary. Oocysts of I. belli are oval and about 32 ×
16 μ in size. Immature oocysts contain a granular
zygote. Mature oocysts contain two sporocysts, each
Fig. 9.11: Cyst and trophozoite of Giardia lamblia with four sporozoites. With modified Ziehl-Neelsen
stain (on a fecal smear prepared from the sediment
after formalin-ether concentration), oocysts stain
Cysts of G. lamblia are 8-12 μ in diameter, oval, uniform red-pink. Under ultraviolet light, oocysts
and contain 4 nuclei, axonemes, median bodies, and show autofluorescence.
remains of flagella (Fig. 9.11).
2. Detection of antigen of G. lamblia in stool sample: Cryptosporidium parvum: Oocysts of C. parvum are
Antigen of G. lamblia can be demonstrated by enzyme difficult to demonstrate in direct fecal wet mounts.
immunoassay technique with high sensitivity (90- They are demonstrated either by modified Ziehl-
99%) and specificity (95-100%). This test can be used Neelsen staining of concentrated fecal smear or by
as the initial test for diagnosis of giardiasis; however, immunofluorescence technique. They are 4-6 μ in size,
stool examination is still important for detection of round to oval, and stain pink-red.
other concomitant parasite infections. Cyclospora cayetanensis: In direct wet mounts of feces,
3. Direct fluorescent antibody assay: This test is oocysts measure 8-10 μ in diameter, and contain a
available commercially in a kit form and is highly cluster of refractile globules (morula-like appearance).
sensitive and specific. Cysts are labeled with With modified Ziehl-Neelsen stain, they appear
immunofluorescent antibodies and are detected similar to C. parvum, but are larger. Under ultraviolet
under fluorescence microscope. light (365 nm), oocysts show intense blue auto-
fluorescence.
Coccidia 2. Detection of antigen in stool samples: Enzyme
Isospora belli, Cryptosporidium parvum, and Cyclospora immunoassay for detection of specific antigen of C.
cayetanensis are human intestinal coccidia. They have a parvum is available. It is more sensitive and specific
worldwide distribution. Transmission is by fecal-oral than Ziehl-Neelsen staining.
route (ingestion of infective oocysts). These protozoan 3. Direct fluorescent antibody assay: This assay is
organisms cause self-limited, mild diarrheal illness; available for Cryptosporidium parvum and is highly
sensitive and specific. Oocysts of Cryptosporidium
however, in immunocompromised patients (such as
labeled with fluorescent antibody are readily detected
patients with acquired immunodeficiency syndrome)
under fluorescence microscope.
they can induce severe and protracted diarrhea, which
may sometimes be life-threatening. Microsporidia
Life cycle: Ingestion of infective oocysts by humans leads Microsporidia are obligate intracellular protozoa, which
to infection. Release of sporozoites from oocysts occurs cause opportunistic infection in immunocompromised
which infect intestinal epithelial cells. Sporozoites patients leading to persistent diarrhea and weight loss.
multiply within epithelial cells with formation of Common species causing infection in humans are
merozoites (asexual reproduction by fission or schizo- Enterocytozoon bieneusi, Encephalitozoon intestinalis, and
gony), which infect other epithelial cells. Some mero- Encephalitozoon hellem.

tahir99 - UnitedVRG
vip.persianss.ir
112 Essentials of Clinical Pathology

Transmission of infection is by ingestion of spores. and intestinal perforation. Sometimes worms can
The organisms develop and multiply in intestinal cells invade pancreatic duct or common bile duct and
and form infective spores; rupture of host cells releases cause obstruction; this can lead to pancreatitis or
spores some of which infect newer cells while others are obstructive jaundice respectively. From the bile duct,
excreted in feces. adult worms can reach liver and cause abscess. Adult
Some microsporidia can cause keratoconjunctivitis, worms can migrate to the appendix to cause
hepatitis, peritonitis, respiratory infection, and kidney appendicitis.
disease. Microsporidia cannot be demonstrated on wet • Malabsorption
mounts because of their very small size.
Laboratory Diagnosis
Laboratory Diagnosis
1. Demonstration of eggs of A. lumbricoides: Diagnosis
1. Modified trichrome stain of stool sample: Spores are
of A. lumbricoides infection is made by demonstration
very small (1-5 μ), stain red, and may show a
of eggs on stool examination. Eggs can be demons-
transverse band.
trated in direct wet mount of feces in moderate to
2. Small intestinal biopsy for demonstration of spores
heavy infections. The recommended procedure is
within intestinal cells.
formol-ethyl acetate sedimentation technique for
concentration of eggs. In feces, four types of eggs are
HELMINTHS found: fertilized (double-shelled or decorticated) and
Ascaris lumbricoides (Roundworm) unfertilised (double-shelled or decorticated).
Fertilized eggs: These are oval, yellow-brown, and
This is the most common helminthic infection of humans.
about 70 μ × 50 μ in size. They have outer and inner
Children are more commonly affected than adults. Mode
shells. Outer shell is uneven, brown (due to staining
of transmission is fecal-oral route (ingestion of infective
by bile), and rough (mamillated), while the inner shell
eggs). Adult worms live in the small intestine (duodenum
is thick, smooth , and colorless. The egg contains a
and jejunum) of the host. Eggs are laid by adult female
single central granular mass (fertilized ovum).
worms (about 200,000 per day), which are excreted in
feces. Eggs can remain viable in soil for many years. Unfertilized eggs: Single female worms discharge these
Contamination can occur when untreated human feces eggs. They are slightly larger and more elongated
are used as a fertilizer or by soiling of hands of playing than the fertilized eggs (90 μ in length). Outer shell is
children. Adult worms can live in the intestine for 1-2 dark brown and more irregular, while the inner shell
years. is thinner. This egg is filled with a mass of large
refractile granules (Fig. 9.12).
Life cycle: Infection is acquired by ingestion of infective
Decorticated eggs do not have the outer uneven shell
eggs via contaminated food or hands. Eggs hatch to
and resemble the hookworm eggs.
release larvae in intestine. Larvae penetrate the mucosa
and enter the bloodstream. Larvae circulate, reach lungs
and penetrate alveolar walls to enter the respiratory tree.
Larvae migrate up the trachea to the epiglottis from
where they are swallowed. Maturation of larvae to adult
worms occurs in the small intestine. Female worms lay
down eggs, which are excreted in feces. Eggs become
embryonated (infective) in 4-6 weeks in favorable
environment.

Clinical Features
• Asymptomatic if infection is light.
• Loeffler’s syndrome: Migration of larvae through the
lungs can induce cough, wheezing, eosinophilia, and
bilateral, irregular pulmonary densities.
• Local effects: These include abdominal pain, diarrhea, Fig. 9.12: Fertilized and unfertilized eggs of
intestinal obstruction due to a large mass of worms, Ascaris lumbricoides

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Feces 113

2. Identification of adult worms: Occasionally adult


worms are passed spontaneously in the feces and
brought to the laboratory for identification. Adult
ascaris worms are cylindrical or round, pinkish, and
measure about 15 cm (male) or 30 cm (female) in
length. Diameter is about 0.5 cm and tail is curved
(male) or straight (female). There are three lips at the
anterior end (mouth).

Hookworms
The hookworms are Ancylostoma duodenale (old world
hookworm) and Necator americanus (new world hook-
worm).
Life cycle: Infection occurs when there is penetration of
skin of foot by filariform larvae present in soil. Larvae Fig. 9.13: Hookworm egg in fresh stools. Egg in oval shape
enter the circulation, and through veins are carried to with a thin shell, has a clear space between wall and developing
the heart and then reach lungs, migrate up the respiratory cleavage, and contains granular cells
tree and are swallowed. Maturation of larvae to the adult
worms occurs in small intestine. Adult worms attach to
the mucosa and suck blood. Adult female worms produce mount of fecal sample can demonstrate eggs in
eggs, which are excreted in feces. Rhabditiform larvae moderate to heavy infections. Eggs of A. duodenale
are released from eggs into the soil and mature into and N. americanus are morphologically similar. They
infective filariform larvae. Both A. duodenale and N. are 50-75 μ in length and 40 μ in width, oval,
americanus infection are acquired when infective colourless, and have a thin shell. In fresh stools, eggs
filariform larvae penetrate the skin. A. duodenale infection show 4-8 gray, granular cells (Fig. 9.13). If stool is
is also acquired by ingestion of infective larvae. more than 12 hours old, eggs will show a rhabditiform
larva folded upon itself. Such egg is called as
Clinical Features
embryonated. If feces are more than 24 hours old, then
Hookworm infection can cause: free rhabditiform larvae will be seen. This should be
• “Ground itch”: This is inflammation and marked differentiated from larvae of Strongyloides stercoralis
itching on the skin at the site of larval penetration. (buccal cavity of hookworm larva is longer).
• Loeffler’s syndrome: This is due to migration of larvae 2. Other laboratory features: Blood examination often
through the lungs. shows eosinophilia. Microcytic hypochromic anemia
• Iron deficiency anemia due to chronic blood loss: This develops due to chronic blood loss. Test for occult
is a well-known and most common complication of blood in stools is positive.
hookworm infection. Adult worms attach themselves
to the small intestinal mucosa by teeth-like structures Trichuris trichiura
or cutting plates, and suck blood. They then change
their sites of attachment (every 4-8 hours) while blood Infection is acquired by ingestion of infective eggs.
continues to ooze from the previous site. One adult Larvae, which are released, develop into adult worms
A. duodenale causes loss of 0.15 ml of blood while one and attach to the mucosa. Released eggs are excreted in
N. americanus causes loss of 0.03 ml per day. feces, and mature in soil to infective stage under suitable
conditions.
• Abdominal pain and diarrhea if worm load is high.
Heavy infection can cause diarrhea with blood and
Laboratory Diagnosis mucus in stools, iron deficiency anaemia, or rectal
prolapse.
1. Demonstration of hookworm eggs: Diagnosis is based Diagnosis depends on identification of typical eggs
on identification of hookworm eggs on stool on stool examination. Eggs measure 50 × 25 μ in size, are
examination. Technique of formol-ethyl acetate yellow-brown and barrel-shaped. A rounded, trans-
sedimentation is preferred; if not available, direct wet parent plug is present at both poles (Fig. 9.14). Eggs

tahir99 - UnitedVRG
vip.persianss.ir
114 Essentials of Clinical Pathology

5. In immunocompromised persons, potentially fatal


hyper-infection (secondary to auto-infection) can
occur causing severe pneumonia, neurologic compli-
cations, abdominal pain, shock, and septicemia.

Laboratory Diagnosis
Identification of larvae of S. stercoralis: Diagnosis of S.
stercoralis infection depends on demonstration of
rhabditiform larvae in fresh stool specimens. Eggs of S.
stercoralis are rarely seen in stool samples because they
hatch and release rhabditiform larvae in the intestine.
Rhabditiform larvae are 200-300 μ in length and 15 μ in
width and are actively motile. They have two esopha-
geal swellings, a prominent genital primordium and a
Fig. 9.14: Egg of Trichuris trichiura in iodine wet mount
short buccal cavity. Sometimes, in old fecal samples,
rhabditiform larvae of hookworms are seen which
resemble those of S. stercoralis; the differentiating feature
contain central, uniformly granular mass. Eggs are often
is the longer buccal cavity of the former.
quantitated to assess the severity of infection.
Excretion of larvae is often irregular and their number
may be few. Therefore, fecal examination may yield
Strongyloides stercoralis
negative result. In suspected cases, concentration
Life cycle: Life cycle is more complex than that of other technique (formol-ethyl acetate) is helpful.
nematodes. Penetration of skin by infective filariform Duodenal fluid can be aspirated for detection of
larvae in soil causes infection. Larvae enter the circulation larvae or Entero-test (String test) can be performed.
and migrate to the lungs, penetrate the alveolar spaces, (Entero-test: A commercially available gelatin capsule
move up the trachea, and are swallowed. Maturation to consists of a textured string. One end of the string is
adult worms occurs in the small intestine. Female worms attached or taped to the cheek and the capsule is then
lay down eggs (parthenogenesis). Eggs hatch to release swallowed. The end of the string reaches and is exposed
rhabditiform larvae in small intestine, which are excreted in the duodenum after several hours. After removal, the
along with feces (Sometimes, rhabditiform larvae can terminal part of the string should be bile-stained. The
mature into filariform larvae, which penetrate mucosa, mucus from the end of the string is wiped onto a glass
or perianal skin and enter the circulation; this is called slide and examined for larvae). In disseminated infection,
as autoinfection). Maturation of rhabditiform larvae to larvae may be detected in sputum.
infective filariform larvae occurs in soil, which can Enzyme immunoassay test that detects IgG antibodies
penetrate the skin and cause infection. If this does not to S. stercoralis is available and is indicated if organism is
happen, larvae develop into adult male and female not detected in feces, duodenal aspirate, or string test
worms which mate and lay down eggs, from which and clinical suspicion is strong. It cannot differentiate
rhabditiform larvae are released which further mature between recent and past infection.
into infective filariform larvae.
Enterobius vermicularis
Clinical Features
E. vermicularis is also called as pinworm, seatworm, or
1. Redness and itching at the site of penetration of oxyuris. It is distributed worldwide. Infection is common
filariform larva. in children. Mode of transmission is ingestion of food or
2. Loeffler’s syndrome due to migration of larva through water contaminated with infective eggs through fingers.
the lungs.
3. Heavy infection can cause abdominal pain, diarrhea, Life cycle: After ingestion, eggs hatch to release larvae in
and malabsorption. the intestine. Larvae mature to the adult worms in the
4. Chronic infection causes abdominal pain, diarrhea, cecum or appendix. Female worms migrate at night to
and urticaria. perianal skin to deposit up to 15,000 eggs. Marked
Examination of Feces 115

irritation at the site leads to contamination of fingers Laboratory Diagnosis


through scratching which causes self-infection by Diagnosis depends on demonstration of eggs in samples
transferring eggs to the mouth. Enterobiasis can also be collected from perianal skin (transparent adhesive tape
acquired by handling contaminated clothes, linen, etc. method) or demonstration of adult worms. Eggs of E.
vermicularis measure 60 μ × 30 μ in size, are oval, and
Clinical Features flattened on one side. They are colorless, transparent with
Intense nocturnal perineal or perianal itching is usual. a double-lined smooth shell, and contain a small granular
Acute appendicitis can occur due to the presence of worm mass (Fig. 9.15) or a larva. Adult pinworms may be
in the appendix. Sometimes, worm can invade female recovered from perianal skin folds (by adhesive tape) or
genital tract leading to vulvovaginitis, and formation of may be found in children’s feces. They are white, motile,
pelvic and peritoneal granulomas. and small in size (male: 0.5 cm; female: 1 cm).

Special technique for collection and demonstration of pinworm Taenia solium


eggs: Adult female pinworm migrates at night from the
intestine (cecum) to the perianal skin and deposits eggs. T. solium occurs mainly in India, Pakistan, China, S.
Pinworm eggs are, therefore, detected in perianal skin Africa, and Latin America. It is transmitted by ingestion
folds and are often not found on routine stool exami- of raw or undercooked pork containing infective
nation. cysticercus larvae (cysticercus cellulosae).
Pinworm eggs can be collected either by a transparent
Life cycle: Infection is acquired by ingestion of raw or
adhesive tape (“cellophane tape test”) or by anal swab.
undercooked pork containing infective encysted
Specimen should preferably be collected late into the
cysticercus larva by man who is a definitive host. Scolex
night or early morning before patient passes urine, feces,
of cysticercus is freed and attaches to the wall of the
or takes a bath.
A transparent adhesive tape is folded over the end intestine by its suckers and hooks. Development of
of a glass slide, spoon handle or a wooden tongue cysticercus into an adult tapeworm occurs by addition
depressor (sticky surface outwards). Patient’s buttocks of multiple segments or proglottids to the scolex. Length
are separated and the slide or spoon handle covered with of the adult tapeworm is about 2-7 meters. Each
tape is pressed over perianal skin at many sites. The tape proglottid is a functional hermaphroditic reproductive
is then spread over a glass slide with adhesive side down unit, which produces numerous eggs. Egg-filled segment
and pressed flat onto the slide surface. Slide covered with at the end of the worm detaches itself from the worm
tape is then examined under the microscope. and releases eggs in feces; segment is also excreted. Eggs
In another method, a cotton swab is rubbed around contaminate the soil and are ingested by the pigs
the anus and then rinsed in a test tube containing 0.5 ml (intermediate hosts) while feeding. Embryo (released
saline. The fluid is drawn in a pipette; a drop is placed from the egg) penetrates intestinal wall of the pig and is
on a glass slide, and observed under the microscope with carried through the bloodstream to the muscles where it
reduced illumination. develops into infective cysticercus larvae.

Fig. 9.15: (A) Enterobius eggs, (B) Enterobius eggs collected by transparent tape method
116 Essentials of Clinical Pathology

Man can become an intermediate host if he eats food


contaminated with eggs or if there is self-contamination
(through contaminated fingers). In such an event, embryo
enters the bloodstream and cysticercus develops at any
body site.

Clinical Features
1. Intestinal infection: Clinical features are usually
insignificant. The patient may notice passage of a flat
segment of the worm in feces.
2. Cysticercosis: This occurs if man accidentally ingests
food contaminated with T. solium eggs (or if there is
auto infection). Nodules containing cysticercus
develop in skeletal muscle, subcutaneous tissue, Fig. 9.16: Egg of Taenia solium/saginata
heart, liver, brain, etc. Cysticercus (or bladder worm)
is a small cyst (< 1.5 cm) containing clear fluid and
inverted scolex. of T. solium shows 4 suckers and a crown of
Involvement of brain (neurocysticercosis) can cause hooklets.
seizures (neurocysticercosis is a common cause of 2. Diagnosis of cysticercosis: Cysticercosis can be
epilepsy in endemic areas in children), raised intracranial diagnosed by serologic tests, radiologic studies, and
tension (due to obstruction to cerebrospinal fluid flow biopsy of the lesion.
by intraventricular cysts), psychiatric disturbances, and Indirect hemagglutination assay has sensitivity of
localizing signs; sometimes sudden death can occur. about 80%. A titer of ≥ 1:64 indicates cysticercosis. Newer
glycoprotein immunoblot assay is more sensitive and
Laboratory Diagnosis specific for diagnosis of neurocysticercosis. .
1. Examination of feces X-ray is helpful in detecting calcified cysts. Computed
tomography scans are helpful in diagnosing neuro-
i. Identification of eggs: Morphologically, eggs of T. cysticercosis.
solium and T. saginata are identical. Distinction
between the two species requires examination of Taenia saginata
proglottids or scolices. Egg measures 30-40 μ in
diameter, is round to oval, and has a thick, brown T. saginata (beef tapeworm) has worldwide distribution
wall with transverse lines. The egg contains an with particularly high prevalence in the Middle East,
embryo, which is a round granular mass contai- Africa, Asia, and Latin America. Mode of transmission
ning 3 pairs of hooklets and surrounded by a fine is eating raw or undercooked beef (containing infective
membrane (Fig. 9.16). Occasionally, the egg is cysticercus bovis larvae).
enclosed in a clear sac. Eggs are discharged Life cycle is similar to that of T. solium except: (i)
intermittently by the tapeworm and therefore may animal host is cattle, (ii) eggs are not infectious to humans
not be detected easily. Repeated stool exami- and therefore human cysticercosis does not occur after
nations and formol-ether concentration technique ingestion of eggs, and (iii) segments of T. saginata also
are often required for their demonstration. migrate to the perianal skin and deposit eggs.
ii. Identification of gravid segments or proglottids: This
allows identification of species. The segment is Clinical Features
flattened between two glass slides and examined
under a magnifying glass. Gravid segment is 13 These are usually insignificant. Sometimes abdominal
mm × 8 mm in size, translucent, and pale blue. It pain and diarrhea can occur.
has a central uterine stem with 8-13 lateral
Laboratory Diagnosis
branches. (Uterine branches are >13 in T. saginata).
iii. Identification of scolex (head): Scolex of a tapeworm 1. Identification of eggs: Eggs of T. saginata can be
is very small (pinhead size) and is rarely seen. identified in feces and perianal skin. They are
When examined with a magnifying glass, scolex morphologically similar to those of T. solium.
Examination of Feces 117

2. Identification of gravid segments: Segments measure 20 • Fecal osmotic gap


mm × 6 mm in size, and are ivory white. They contain • Fecal pH
a central uterine stem with more than 13 side branches
Test for Occult Blood in Stools
(i.e. 15-20 branches).
3. Identification of head or scolex: Scolex of T. saginata has Presence of blood in feces which is not apparent on gross
4 suckers but no hooklets. It measures 2 mm in width. inspection and which can be detected only by chemical
Various laboratory tests for diagnosis of parasitic tests is called as occult blood. Causes of occult blood in
infection of intestine are summarized in Table 9.3. stools are:
i. Intestinal diseases: hookworms, amebiasis,
CHEMICAL EXAMINATION typhoid fever, ulcerative colitis, intussusception,
adenoma, cancer of colon or rectum.
Chemical examination of feces is usually carried out for ii. Gastric and esophageal diseases: peptic ulcer,
the following tests (Fig. 9.17): gastritis, esophageal varices, hiatus hernia.
• Occult blood iii. Systemic disorders: bleeding diathesis, uremia.
• Excess fat excretion (malabsorption) iv. Long distance runners.
• Urobilinogen Occult blood test is recommended as a screening
• Reducing sugars procedure for detection of asymptomatic colorectal

Fig. 9.17: Chemical examinations done on fecal sample

Table 9.3: Summary of laboratory tests for diagnosis of intestinal parasites


Test Use

1. Direct saline mount Trophozoites, cysts, ova, and larvae


2. Direct iodine mount Protozoal cysts
3. Faecal concentration Recovery of protozoal cysts, helminth ova and larvae
4. Cellophane tape technique Ova of Enterobius vermicularis
5. Trichrome stain of fecal smear Protozoal cysts and trophozoites
6. AFB stain of fecal smear Oocysts of Cryptosporidium, Cyclospora, and Isospora
7. Detection of antigen in random stool sample E. histolytica, G. lamblia, Cryptosporidium
8. Molecular methods based on polymerase E. histolytica, G. lamblia, Cryptosporidium, Microsporidia,
chain reaction on stool sample Cyclospora
9. Serologic methods E. histolytica, S. stercoralis, Cysticercus
118 Essentials of Clinical Pathology

cancer. Yearly examinations should be carried out after is measured in fecal sample and in simultaneously
the age of 50 years. If the test is positive, endoscopy and collected blood specimen. Radioactivity in feces indicates
barium enema are indicated. gastrointestinal bleeding. Amount of blood loss can be
calculated. Although the test is sensitive, it is not suitable
Tests for detection of occult blood in feces: Many tests are
available which differ in their specificity and sensitivity. for routine screening.
These tests include tests based on peroxidase-like activity Apt test: This test is done to decide whether blood in the
of hemoglobin (benzidine, orthotolidine, aminophena- vomitus or in the feces of a neonate represents swallowed
zone, guaiac), immunochemical tests, and radioisotope maternal blood or is the result of bleeding in the
tests. gastrointestinal tract. The test was devised by Dr. Apt
and hence the name. The baby swallows blood during
Tests Based on Peroxidase-like
delivery or during breastfeeding if nipples are cracked.
Activity of Hemoglobin
Apt test is based on the principle that if blood is of
Principle: Hemoglobin has peroxidase-like activity and neonatal origin it will contain high proportion of
releases oxygen from hydrogen peroxide. Oxygen hemoglobin F (Hb F) that is resistant to alkali denatu-
molecule then oxidizes the chemical reagent (benzidine, ration. On the other hand, maternal blood mostly
orthotolidine, aminophenazone, or guaiac) to produce a contains adult hemoglobin or Hb A that is less resistant.
colored reaction product.
Benzidine and orthotolidine are carcinogenic and are Test for Malabsorption of Fat
no longer used. Benzidine test is also highly sensitive
and false-positive reactions are common. Dietary fat is absorbed in the small intestine with the
Since bleeding from the lesion may be intermittent, help of bile salts and pancreatic lipase. Fecal fat mainly
repeated testing may be required. consists of neutral fats (unsplit fats), fatty acids, and soaps
(fatty acid salts). Normally very little fat is excreted in
Causes of False-positive Tests feces (<7 grams/day in adults). Excess excretion of fecal
fat indicates malabsorption and is known as steatorrhea.
1. Ingestion of peroxidase-containing foods like red It manifests as bulky, frothy, and foul-smelling stools,
meat, fish, poultry, turnips, horseradish, cauliflower, which float on the surface of water.
spinach, or cucumber. Diet should be free from
peroxidase-containing foods for at least 3 days prior Causes of Malabsorption of Fat
to testing.
2. Drugs like aspirin and other anti-inflammatory drugs, i. Deficiency of pancreatic lipase (insufficient
which increase blood loss from gastrointestinal tract lipolysis): chronic pancreatitis, cystic fibrosis.
in normal persons. ii. Deficiency of bile salts (insufficient emulsification
of fat): biliary obstruction, severe liver disease, bile
Causes of False-negative Tests salt deconjugation due to bacterial overgrowth in
1. Foods containing large amounts of vitamin C. the small intestine.
2. Conversion of all hemoglobin to acid hematin (which iii. Diseases of small intestine: tropical sprue, celiac
has no peroxidase-like activity) during passage disease, Whipple’s disease.
through the gastrointestinal tract. Tests for fecal fat are qualitative (i.e. direct micro-
scopic examination after fat staining), and quantitative
Immunochemical Tests (i.e. estimation of fat by gravimetric or titrimetric
These tests specifically detect human hemoglobin. analysis).
Therefore there is no interference from animal hemo- 1. Microscopic stool examination after staining for fat:
globin or myoglobin (e.g. meat) or peroxidase-containing A random specimen of stool is collected after putting
vegetables in the diet. the patient on a diet of >80 gm fat per day. Stool
The test consists of mixing the sample with latex sample is stained with a fat stain (oil red O, Sudan
particles coated with anti-human haemoglobin antibody, III, or Sudan IV) and observed under the microscope
and if agglutination occurs, test is positive. This test can for fat globules (Fig. 9.18). Presence of ≥60 fat
detect 0.6 ml of blood per 100 grams of feces. droplets/HPF indicates steatorrhea. Ingestion of
mineral or castor oil and use of rectal suppositories
51
Radioisotope Test Using Cr can cause problems in interpretation.
In this test, 10 ml of patient’s blood is withdrawn, labeled 2. Quantitative estimation of fecal fat: The definitive
with 51Cr, and re-infused intravenously. Radioactivity test for diagnosis of fat malabsorption is quantitation
Examination of Feces 119

tract obstruction, severe liver disease, oral antibiotic


therapy (disturbance of intestinal bacterial flora), and
aplastic anemia (low hemoglobin turnover). Stools
become pale or clay-colored if urobilinogen is reduced
or absent.

Test for Reducing Sugars


Deficiency of intestinal enzyme lactase is a common
cause of malabsorption. Lactase converts lactose (in milk)
to glucose and galactose. If lactase is deficient, lactose is
converted to lactic acid with production of gas. In infants
this leads to diarrhea, vomiting, and failure to thrive.
Benedict’s test or ClinitestTM tablet test for reducing
sugars is used to test freshly collected stool sample for
lactose. In addition, oral lactose tolerance test is abnormal
(after oral lactose, blood glucose fails to rise above 20
mg/dl of basal value) in lactase deficiency. Rise in blood
glucose indicates that lactose has been hydrolysed and
Fig. 9.18: Sudan stain on fecal sample: (A) Negative; (B) Positive absorbed by the mucosa. Lactose tolerance test is now
replaced by lactose breath hydrogen testing. In lactase
deficiency, accumulated lactose in the colon is rapidly
of fecal fat. Patient should be on a diet of 70-100 gm fermented to organic acids and gases like hydrogen.
of fat per day for 6 days before the test. Feces are Hydrogen is absorbed and then excreted through the
collected over 72 hours and stored in a refrigerator
lungs into the breath. Amount of hydrogen is then
during the collection period. Specimen should not be
measured in breath; breath hydrogen more than 20 ppm
contaminated with urine. Fat quantitation can be
above baseline within 4 hours indicates positive test.
done by gravimetric or titrimetric method. In
gravimetric method, an accurately weighed sample
Fecal Osmotic Gap
of feces is emulsified, acidified, and fat is extracted
in a solvent; after evaporation of solvent, fat is Fecal osmotic gap is calculated from concentration of
weighed as a pure compound. Titrimetric analysis is electrolytes in stool water by formula 290-2([Na+] + [K+]).
the most widely used method. An accurately weighed (290 is the assumed plasma osmolality). In osmotic
stool sample is treated with alcoholic potassium diarrheas, osmotic gap is >150 mOsm/kg, while in
hydroxide to convert fat into soaps. Soaps are then secretory diarrhea, it is typically below 50 mOsm/kg.
converted to fatty acids by the addition of hydro- Evaluation of chronic diarrhea is shown in Figure 9.19.
chloric acid. Fatty acids are extracted in a solvent and
the solvent is evaporated. The solution of fat made in Fecal pH
neutral alcohol is then titrated against sodium Stool pH below 5.6 is characteristic of carbohydrate
hydroxide. Fatty acids comprise about 80% of fecal malabsorption.
fat. Values >7 grams/day are usually abnormal.
Values >14 grams/day are specific for diseases REFERENCE RANGES
causing fat malabsorption.
Bulk: 100-200 grams/day
Test for Urobilinogen in Feces Color: Brown
Fecal urobilinogen is determined by Ehrlich’s aldehyde Water: Up to 75%
test (see Chapter 1 “Examination of Urine”). Specimen pH: 7.0-7.5
should be fresh and kept protected from light. Normal Red blood cells: Absent
amount of urobilinogen excreted in feces is 50-300 mg White blood cells: Few
per day. Increased fecal excretion of urobilinogen is seen Epithelial cells: Present
in hemolytic anemia. Urobilinogen is deceased in biliary Crystals: Calcium oxalate, triple phosphate
120 Essentials of Clinical Pathology

Fig. 9.19: Evaluation of chronic diarrhea

Fat (Adults): <7 grams/day (gravimetric method), <6 BIBLIOGRAPHY


grams/day (titrimetric method) 1. American Gastroenterological Association. AGA
Fat droplets: Average 2.5 per high power field in random technical review on the evaluation and management
sample of chronic diarrhea. Gastroenterology 1999;116:
1464-86.
Urobilinogen: 50-300 mg/24 hours 2. American Gastroenterological Association Medical
Parasites: Nil Position Statement: Guidelines for the evaluation and
management of chronic diarrhoea. Gastroenterology
Ova, cysts, trophozoites: Nil 1999;116:1461-3.
3. Haque R, Huston CD, Hughes M, Houpt E, Petri, WA
Critical Values Jr. Amebiasis. New Engl J Med 2003;348:1565:73.
Stool culture positive for Salmonella, Shigella, Campylo- 4. Kucik CJ, Martin GL, Sortor BV. Common intestinal
bacter, Vibrio, or Yersinia. parasites. Am Fam Physician 2004;69:1161-8.
10

Gastric Analysis

Gastric analysis consists of quantitation of gastric acid into pyloric antrum and pyloric canal. It is lined by
(basal and pentagastrin-stimulated) produced by the mucus-secreting cells and gastrin-secreting neuro-
stomach. Gastric juice is collected by a nasogastric tube endocrine cells (G cells) (Fig. 10.1).
and gastric acid is quantitated by its titration with sodium In the stomach, ingested food is mechanically and
hydroxide solution. chemically broken down to form semi-digested liquid
called chyme. Following relaxation of pyloric sphincter,
NORMAL GASTRIC ANATOMY AND chyme passes into the duodenum.
PHYSIOLOGY There are three phases of gastric acid secretion:
cephalic, gastric, and intestinal.
Anatomically, stomach is divided into four parts: cardia, • Cephalic or neurogenic phase: This phase is initiated
fundus, body, and pyloric part. Cardia is the upper part by the sight, smell, taste, or thought of food that causes
surrounding the entrance of the esophagus and is lined stimulation of vagal nuclei in the brain. Vagus nerve
by the mucus-secreting epithelium. The epithelium of directly stimulates parietal cells to secrete acid; in
the fundus and the body of the stomach is composed of addition, it also stimulates antral G cells to secrete
different cell types including: (i) mucus-secreting cells gastrin in blood (which is also a potent stimulus for
which protect gastric mucosa from self-digestion by gastric acid secretion) (Fig. 10.2). Cephalic phase is
forming an overlying thick layer of mucus, (ii) parietal abolished by vagotomy.
cells which secrete hydrochloric acid and intrinsic factor, • Gastric phase: Entry of swallowed food into the
and (iii) peptic cells or chief cells which secrete the stomach causes gastric distension and induces gastric
proteolytic enzyme pepsinogen. Pyloric part is divided phase. Distension of antrum and increase in pH due

Fig. 10.1: Parts of stomach and their lining cells


122 Essentials of Clinical Pathology

Fig. 10.2: Stimulation of gastric acid secretion. Three receptors on parietal cells stimulate acid secretion: histamine (H2) receptor,
acetylcholine or cholinergic receptor, and gastrin/CCK-B receptor. Histamine is released by enterochromaffin-like cells in lamina
propria. Acetylcholine is released from nerve endings. Gastrin is released from G cells in antrum (in response to amino acids
in food, antral distention, and gastrin-releasing peptide). After binding to receptors, H+ is secreted in exchange for K+ by
proton pump

to neutralization of acid by food stimulate antral G the formation of large polypeptide molecules (optimal
cells to secrete gastrin into the circulation. Gastrin, in function at pH 1.0 to 3.0). Its secretion is enhanced by
turn, causes release of hydrochloric acid from parietal vagal stimulation.
cells. • Mucus
• Intestinal phase: Entry of digested proteins into the • Intrinsic factor (IF): IF is necessary for absorption of
duodenum causes an increase in acid output from vitamin B12 in the terminal ileum. It is secreted by
the stomach. It is thought that certain hormones and parietal cells of stomach.
absorbed amino acids stimulate parietal cells to
secrete acid. INDICATIONS FOR GASTRIC ANALYSIS
The secretion from the stomach is called as gastric juice. In gastric analysis, amount of acid secreted by the
The chief constituents of the gastric juice are: stomach is determined on aspirated gastric juice sample.
• Hydrochloric acid (HCl): This is secreted by the parietal Gastric acid output is estimated before and after
cells of the fundus and the body of the stomach. HCl stimulation of parietal cells (i.e. basal and peak acid
provides the high acidic pH necessary for activation output). This test was introduced in the past mainly for
of pepsinogen to pepsin. Gastric acid secretion is the evaluation of peptic ulcer disease (to assess the need
stimulated by histamine, acetylcholine, and gastrin for operative intervention). However, diminishing
(Fig. 10.2). HCl kills most microorganisms entering frequency of peptic ulcer disease and availability of safe
the stomach and also denatures proteins (breaks and effective medical treatment have markedly reduced
hydrogen bonds making polypeptide chains to the role of surgery.
unfold). Its secretion is inhibited by somatostatin 1. To determine the cause of recurrent peptic ulcer
(secreted by D cells in pancreas and by mucosa of disease:
intestine), gastric inhibitory peptide (secreted by K • To detect Zollinger-Ellison (ZE) syndrome: ZE
cells in duodenum and jejunum), prostaglandin, and syndrome is a rare disorder in which multiple
secretin (secreted by S cells in duodenum). mucosal ulcers develop in the stomach, duode-
• Pepsin: Pepsin is secreted by chief cells in stomach. num, and upper jejunum due to gross hyper-
Pepsin causes partial digestion of proteins leading to secretion of acid in the stomach. The cause of
Gastric Analysis 123

excess secretion of acid is a gastrin-producing congestive cardiac failure, or non-cooperative patient.


tumor of pancreas. Gastric analysis is done to • Pyloric stenosis: Obstruction of gastric outlet can
detect markedly increased basal and pentagastrin- elevate gastric acid output due to raised gastrin
stimulated gastric acid output for diagnosis of ZE (following antral distension).
syndrome (and also to determine response to acid- • Pentagastrin stimulation is contraindicated in cases
suppressant therapy). However, a more sensitive with allergy to pentagastrin, and recent severe gastric
and specific test for diagnosis of ZE syndrome is hemorrhge due to peptic ulcer disease.
measurement of serum gastrin (fasting and
secretin-stimulated). Gastric analysis is not a commonly performed proce-
• To decide about completeness of vagotomy dure because of following reasons:
following surgery for peptic ulcer disease: See • It is an invasive and cumbersome technique that is
Hollander’s test, later. traumatic and unpleasant for the patient.
2. To determine the cause of raised fasting serum • Information obtained is not diagnostic in itself.
gastrin level: Hypergastrinemia can occur in • Availability of better tests for diagnosis such as
achlorhydria, Zollinger-Ellison syndrome, and antral endoscopy and radiology (for suspected peptic ulcer
G cell hyperplasia. or malignancy); serum gastrin estimation (for ZE
3. To support the diagnosis of pernicious anemia (PA): syndrome); vitamin assays, Schilling test, and anti-
Pernicious anemia is caused by defective absorption parietal cell antibodies (for pernicious anemia); and
of vitamin B12 due to failure of synthesis of intrinsic tests for Helicobacter pylori infection (in duodenal or
factor secondary to gastric mucosal atrophy. There is gastric ulcer).
also absence of hydrochloric acid in the gastric juice • Availability of better medical line of treatment that
(achlorhydria). Gastric analysis is done for demons- obviates need for surgery in many patients.
tration of achlorhydria if facilities for vitamin assays
and Schilling’s test are not available (Achlorhydria Method of Gastric Analysis
by itself is insufficient for diagnosis of PA).
To assess gastric acid secretion, acid output from the
4. To distinguish between benign and malignant
stomach is measured in a fasting state and after injection
ulcer: Hypersecretion of acid is a feature of duodenal
of a drug which stimulates gastric acid secretion.
peptic ulcer, while failure of acid secretion
Basal acid output (BAO) is the amount of hydro-
(achlorhydria) occurs in gastric carcinoma. However,
anacidity occurs only in a small proportion of cases chloric acid (HCl) secreted in the absence of any external
with advanced gastric cancer. Also, not all patients stimuli (visual, olfactory, or auditory).
with duodenal ulcer show increased acid output. Maximum acid output (MAO) is the amount of
5. To measure the amount of acid secreted in a patient hydrochloric acid secreted by the stomach following
with symptoms of peptic ulcer dyspepsia but stimulation by pentagastrin. MAO is calculated from the
normal X-ray findings: Excess acid secretion in such first four 15-minute samples after stimulation.
cases is indicative of duodenal ulcer. However, Peak acid output (PAO) is calculated from the two
hypersecretion of acid does not always occur in highest consecutive 15-minute samples. It indicates
duodenal ulcer. greatest possible acid secretory capacity and is preferred
6. To decide the type of surgery to be performed in a over MAO as it is more reproducible.
patient with peptic ulcer: Raised basal as well as peak Acidity is estimated by titration (see later).
acid outputs indicate increased parietal cell mass and
need for gastrectomy. Raised basal acid output with Collection of Sample
normal peak output is an indication for vagotomy.
All drugs that affect gastric acid secretion (e.g. antacids,
anticholinergics, cholinergics, H2-receptor antagonists,
CONTRAINDICATIONS TO GASTRIC
antihistamines, tranquilizers, antidepressants, and
ANALYSIS
carbonic anhydrase inhibitors) should be withheld for
• Gastric intubation for gastric analysis is contra- 24 hours before the test. Proton pump inhibitors should
indicated in esophageal stricture or varices, active be discontinued 5 days prior to the test. Patient should
nasopharyngeal disease, diverticula, malignancy, be relaxed and free from all sources of sensory stimula-
recent history of severe gastric hemorrhage, hyper- tion.
tension, aortic aneurysm, cardiac arrhythmias, Patient should drink or eat nothing after midnight.
124 Essentials of Clinical Pathology

Gastric juice can be aspirated through an oral or that has accumulated overnight is aspirated and
nasogastric tube (polyvinyl chloride, silicone, or discarded. This is followed by aspiration of gastric
polyurethane) or during endoscopy. secretions at 15-minute intervals for 1 hour (i.e. total 4
Oral or nasogastric tube (Fig. 10.3) is commonly used. consecutive samples are collected). All the samples are
It is a flexible tube having a small diameter and a bulbous centrifuged to remove any particulate matter. Each 15-
end that is made heavy by a small weight of lead. The minute sample is analyzed for volume, pH, and acidity.
end is perforated with small holes to allow entry of gastric The acid output in the four samples is totaled and the
juice into the tube. As the end is radiopaque, the tube result is expressed as concentration of acid in milliequi-
can be positioned in the most dependent part of the valents per hour or in mmol per hour.
stomach under fluoroscopic or X-ray guidance. The tube After the collection of gastric juice for determination
is lubricated and can be introduced either through the of BAO, patient is given a subcutaneous or intramuscular
mouth or the nose. The patient is either sitting or reclining injection of pentagastrin (6 μg/kg of body weight), and
on left side. The tube has three or four markings on its immediately afterwards, gastric secretions are aspirated
outer surface that correspond with distance of the tip of at 15-minute intervals for 1 hour (for estimation of MAO
the tube from the teeth, i.e. 40 cm (tip to cardio- or PAO). MAO is calculated from the first four 15-minute
esophageal junction), 50 cm (body of stomach), 57 cm samples after stimulation. PAO is calculated from two
(pyloric antrum), and 65 cm (duodenum). The position consecutive 15-minute samples showing highest acidity.
of the tube can be verified either by fluoroscope or by
‘water recovery test’. In the latter test, 50 ml of water is Titration
introduced through the tube and aspirated again;
Gastric acidity is estimated by titration, with the end
recovery of > 90% of water is indicative of proper
point being determined either by noting the change in
placement. The tube is usually positioned in the antrum.
color of the indicator solution or till the desired pH is
A syringe is attached to the outer end of the tube for the
reached.
aspiration of gastric juice.
For estimation of BAO, sample is collected in the In titration, a solution of alkali (0.1 N sodium
morning after 12-hour overnight fast. Gastric secretion hydroxide) is added from a graduated vessel (burette)
to a known volume of acid (i.e. gastric juice) till the end
point or equivalence point of reaction is reached. The
concentration of acid is then determined from the
concentration and volume of alkali required to neutralize
the particular volume of gastric juice. Concentration of
acid is expressed in terms of milliequivalents per liter or
mmol per liter.
Free acidity refers to the concentration of HCl present
in a free, uncombined form in a solution. The volume of
alkali added to the gastric juice till the Topfer’s reagent
(an indicator added earlier to the gastric juice) changes
color or when the pH (as measured by the pH meter)
reaches 3.5 is a measure of free acidity. A screening test
can be carried out for the presence of free HCl in the
gastric juice. If red color develops after addition of a drop
of Topfer’s reagent to an aliquot of gastric juice, free HCl
is present and the diagnosis of pernicious anaemia
(achlorhydria) can be excluded.
Combined acidity refers to the amount of HCl
combined with proteins and mucin and also includes
Fig. 10.3: Oral or nasogastric Ryle’s tube. The tube is marked small amount of weak acids present in gastric juice.
at 40, 50, 57, and 65 cm with radiopaque lines for accurate
Total acidity is the sum of free and combined acidity.
placement. The tip is bulbous and contains a small weight
of lead to assist the passage during intubation and to know
The amount of alkali added to the gastric juice till
the position under fluoroscopy or X-ray guidance. There are phenolphthalein indicator (added earlier to the gastric
four perforations or eyes to aspirate contents from the stomach juice) changes color is a measure of total acidity
through a syringe attached to the base (Box 10.1).
Gastric Analysis 125

5. Peak acid output:


Box 10.1: Determination of basal acid output, maximum
acid output, and peak acid output • Normal: 1-20 mEq/hour.
• Duodenal ulcer: 20-60 mEq/hour.
• Basal acid output (BAO)= Total acid content in all four • Zollinger-Ellison syndrome: > 60 mEq/hour.
15-minute basal samples in mEq/L • Achlorhydria: 0 mEq/hour.
• Maximum acid output (MAO) = Total acid content in all Normal PAO is seen in gastric ulcer and gastric
four 15-minute post-pentagastrin samples in mEq/L carcinoma. Values up to 60 mEq/hour can occur in some
• Peak acid output (PAO) = Sum of two consecutive 15- normal individuals and in some patients with Zollinger-
minute post-pentagastrin samples showing highest acidity Ellison syndrome.
×2 (mEq/L) In pernicious anemia, there is no acid output due to
gastric mucosal atrophy. Achlorhydria should be
diagnosed only if there is no free HCl even after
maximum stimulation.
Interpretation of Results
6. Ratio of basal acid output to peak acid output (BAO/
1. Volume: Normal total volume is 20-100 ml (usually PAO):
< 50 ml). Causes of increased volume of gastric juice • Normal: < 0.20 (or < 20%).
are— • Gastric or duodenal ulcer: 0.20-0.40 (20-40%).
• Delayed emptying of stomach: pyloric stenosis • Duodenal ulcer: 0.40-0.60 (40-60%).
• Increased gastric secretion: duodenal ulcer, • Zollinger-Ellison syndrome: > 0.60 (> 60%).
Zollinger-Ellison syndrome. Normal values occur in gastric ulcer or gastric
2. Color: Normal gastric secretion is colorless, with a carcinoma.
Conditions associated with change in gastric acid
faintly pungent odor. Fresh blood (due to trauma, or
output are listed in Table 10.1.
recent bleeding from ulcer or cancer) is red in color.
It is to be noted that values of acid output are not
Old hemorrhage produces a brown, coffee-ground
diagnostic by themselves and should be correlated with
like appearance (due to formation of acid hematin).
clinical, radiological, and endoscopic features.
Bile regurgitation produces a yellow or green color.
3. pH: Normal pH is 1.5 to 3.5. In pernicious anemia,
pH is greater than 7.0 due to absence of HCl. OTHER TESTS FOR GASTRIC ANALYSIS
4. Basal acid output: 1. Hollander’s test (Insulin hypoglycemia test): In the
• Normal: Up to 5 mEq/hour. past, this test was used for confirmation of complete-
• Duodenal ulcer: 5-15 mEq/hour. ness of vagotomy (done for duodenal ulcer).
• Zollinger-Ellison syndrome: >20 mEq/hour. Hypoglycemia is a potent stimulus for gastric acid
Normal BAO is seen in gastric ulcer and in some secretion and is mediated by vagus nerve. This
patients with duodenal ulcer. response is abolished by vagotomy.

Table 10.1: Causes of alterations in gastric acid output

Increased gastric acid output Decreased gastric acid output

• Duodenal ulcer • Chronic atrophic gastritis


• Zollinger-Ellison syndrome 1. Pernicious anemia
• Hyperplasia of antral G cells 2. Rheumatoid arthritis
• Systemic mastocytosis 3. Thyrotoxicosis
• Basophilic leukemia • Gastric ulcer
• Gastric carcinoma
• Chronic renal failure
• Post-vagotomy
• Post-antrectomy
126 Essentials of Clinical Pathology

In this test, after determining BAO, insulin is results may be obtained. The test is no longer in use.
administered intravenously (0.15-0.2 units/kg) and 4. Spot check of gastric pH: According to some
acid output is estimated every 15 minutes for 2 hours investigators, spot determination of pH of fasting
(8 post-stimulation samples). Vagotomy is considered gastric juice (obtained by nasogastric intubation) can
as complete if, after insulin-induced hypoglycemia detect the presence of hypochlorhydria (if pH>5.0 in
(blood glucose < 45 mg/dl), no acid output is men or >7.0 in women).
5. Congo red test during esophagogastroduodeno-
observed within 45 minutres.
scopy: This test is done to determine the completeness
The test gives reliable results only if blood glucose
of vagotomy. Congo red dye is sprayed into the
level falls below 50 mg/dl at some time following stomach during esophagogastroduodenoscopy; if it
insulin injection. It is best carried out after 3-6 months turns red, it indicates presence of functional parietal
of vagotomy. cells in stomach with capacity of producing acid.
The test is no longer recommended because of the
risk associated with hypoglycemia. Myocardial REFERENCE RANGES
infarction, shock, and death have also been reported. • Volume of gastric juice: 20-100 ml
2. Fractional test meal: In the past, test meals (e.g. oat • Appearance: Clear
meal gruel, alcohol) were administered orally to • pH: 1.5 to 3.5
stimulate gastric secretion and determine MAO or • Basal acid output: Up to 5 mEq/hour
PAO. Currently, parenteral pentagastrin is the gastric • Peak acid output: 1 to 20 mEq/hour
stimulant of choice. • Ratio of basal acid output to peak acid output: <0.20
3. Tubeless gastric analysis: This is an indirect and or < 20%
rapid method for determining output of free
BIBLIOGRAPHY
hydrochloric acid in gastric juice. In this test, a cation-
exchange resin tagged to a dye (azure A) is orally 1. Burtis CA, Ashwood ER (Eds). Tietz Fundamentals of
Clinical Chemistry, 4th ed. Philadelphia: WB Saunders
administered. In the stomach, the dye is displaced
Co, 1996.
from the resin by the free hydrogen ions of the 2. Drossman DA, Shaheen NJ, Grimm IS (Eds). Handbook
hydrochloric acid. The displaced azure A is absorbed of Gastroenterologic Procedures (4th Ed). Philadelphia:
in the small intestine, enters the bloodstream, and is Lippincott Williams and Wilkins, 2005.
excreted in urine. Urinary concentration of the dye is 3. Rosenfeld L. Gastric tubes, meals, acid, and analysis-
rise and decline. Clin Chem 1997;43:837-42.
measured photometrically or by visual comparison
4. Wallach J. Interpretation of Diagnostic tests (7th Ed).
with known color standards. The quantity of the dye Philadelphia. Lippincott: Williams and Wilkins, 2000.
excreted is proportional to the gastric acid output. 5. Wolfe MM, Soll AH. The physiology of gastric acid
However, if kidney or liver function is impaired, false secretion. N Engl J Med 1988;319:1707-14.
11

Tests for Malabsorption and


Pancreatic Function

MALABSORPTION SYNDROMES gastrointestinal tract. Causes of malabsorption are listed


in Table 11.1.
Normal Absorption of Carbohydrates,
Fats, and Proteins Evaluation of Malabsorption Syndromes
Normal digestive processes of carbohydrates, fats, and Clinical Features
proteins are presented in Figures 11.1 to 11.3. The manifestations of malabsorption include diarrhea
(of osmotic type as non-absorption of solutes increases
Causes of Malabsorption
intraluminal osmotic pressure and outpouring of water),
Malabsorption is a term that includes disorders that cause steatorrhea (passage of frothy, bulky, and malodorous
deficient digestion or absorption of nutrients in the stools that are difficult to flush, with oil droplets floating

Fig. 11.1: Carbohydrate digestion and absorption. Carbohydrate digestion and absorption depend mainly on normal
pancreatic function (amylase) and normal intestinal mucosal cells (disaccharidases)
128 Essentials of Clinical Pathology

Fig. 11.2: Digestion and absorption of fat. This requires presence of normal functioning and
adequate absorptive area of intestinal mucosa, bile salts, and pancreatic enzymes

Table 11.1: Causes of malabsorption

Impaired digestion
1. Diseases of pancreas: Chronic pancreatitis, cystic fibrosis,
resection of pancreas
2. Deficiency of bile salts: Impaired excretion of bile
(cholestasis), blind loop syndrome (bile salt deconjugation),
intestinal resection (impaired enterohepatic circulation)

Impaired absorption
1. Disorders of mucosa: Tropical sprue, giardiasis, celiac
disease, Crohn’s disease, Whipple’s disease, radiation
enteritis, amyloidosis, disaccharidase deficiency (e.g.
lactase), abetalipoproteinemia
2. Intestinal resection
3. Lymphatic obstruction: Lymphoma

on toilet water; results from malabsorption of fat), or


deficiency of nutrients (e.g. iron, vitamin B12, folate,
vitamin K, vitamin D, calcium and electrolytes leading
to anemia, bleeding, bone pain, neurological manifes-
tations, etc.). Other manifestations include peripheral
edema, abdominal distention, weight loss and fatigue.
Fig. 11.3: Protein digestion and absorption. Proteolytic enzymes Laboratory Evaluation
of pancreas are trypsin, chymotrypsin, elastase, and carboxy-
peptidase. Protein absorption mainly depends on pancreatic A variety of laboratory tests are available for assessment
proteolytic enzymes and small intestinal cells (enterocytes) of a patient presenting with clinical manifestations of
Tests for Malabsorption and Pancreatic Function 129

malabsorption. In the beginning, complete blood count stain) is poorly validated and often misses mild forms
(for detecting anemia); iron studies, vitamin B12, and of steatorrhoea (See Chapter 9 “Examination of
folate levels; serum albumin, and serum calcium; Feces”).
erythrocyte sedimentation rate (raised in Crohn’s disease 2. Butter fat test: In this test, a standard amount of fat
and Whipple’s disease), and prothrombin time (for is ingested and if chylomicrons are observed in blood
malabsorption of vitamin K) should be obtained. after the fatty meal, fat digestion and absorption are
considered to be adequate.
Radiological investigations (such as small bowel barium
3. Estimation of fecal fat: Patient should take 70 g per
enema, CT scan of abdomen, plain X-ray film of
day fat diet for at least 6 days. This is followed by
abdomen, etc.) can be helpful in certain cases. collection of feces for atleast 3 days and measurement
Laboratory tests for malabsorption can be classified of fecal mass or volume and fat. Fecal fat excretion >7%
as follows: is considered as abnormal. The test is unpleasant to
• Tests for malabsorption of fat perform and misleading results are often obtained due
• Tests for malabsorption of carbohydrates to incompleteness of collection and poor analytical
• Tests for pancreatic function performance (See Chapter 9 “Examination of feces”).
• Tests for malabsorption of vitamin B12 (See ‘Schilling 4. 14CO2-breath tests: Labeled triglycerides (e.g. 14C-
test’ in Chapter on ‘Approach to Diagnosis of triolein) are administered orally; after hydrolysis by
Anemias’) pancreatic lipase to monoglycerides and free fatty
• Tests for bacterial overgrowth in small intestine acids, micelles are formed with the aid of bile acids
• Tests for laxative abuse. that are then absorbed. Ultimately metabolism leads
Tests for malabsorption of fat These tests include: to their excretion as CO2. Measurement of breath 14CO2
• Microscopic examination of feces for fat should show peak radioactivity at 5 to 7 hours. Result
• Butter fat test depends both on fat digestion and absorption. To
• Estimation of fecal fat distinguish between the two, the test is repeated after
• 14CO2 breath tests, e.g. 14C-triolein breath test. administration of a radiolabeled free fatty acid. This
1. Microscopic examination for fecal fat: This test (that test is indirect and qualitative rather than quantitative.
detects fat globules in feces after staining with a fat Evaluation of steatorrhea is shown in Figure 11.4.

Fig. 11.4: Evaluation of steatorrhea or malabsorption of fat


130 Essentials of Clinical Pathology

Tests for malabsorption of carbohydrates the large intestine, where hydrogen is produced by
These tests include: bacterial fermentation (Note: Hydrogen cannot be
• Lactose tolerance test produced by mammalian cells and bacterial fermen-
• Breath hydrogen test tation of unabsorbed carbohydrate is responsible for
• D-xylose absorption test. its presence in the expired air). About 20% of such
1. Lactose tolerance test: Lactose is a carbohydrate hydrogen is absorbed by mucosa and excreted
consisting of one molecule of glucose and one through respiration. In lactase deficiency, breath
molecule of galactose. It is also called as milk sugar hydrogen is increased following lactose load.
as it comprises of 2-8% of milk. A 50 g oral dose of Clinically such patients also develop symptoms of
lactose in 400 ml of water is given and blood glucose lactose intolerance such as abdominal distention,
response is measured as in oral glucose tolerance test discomfort, flatulence and diarrhea.
Lactase deficiency may be congenital or acquired.
(at 0, 30, 60, and 120 minutes) and patient is also
Transient deficiency occurs following damage to gut
observed for development of symptoms. Failure of
mucosa, e.g. gastroenteritis, celiac disease, and
expected rise of blood glucose (i.e. peak rise of blood
inflammatory bowel disease.
glucose <20 mg/dl) and provocation of symptoms
3. D-xylose absorption test: This test assesses the
are indicative of lactose intolerance (Fig. 11.5). To
absorptive capacity of small intestinal mucosa. D-
improve specificity, test is repeated following xylose, a plant sugar, is completely absorbed in the
administration of glucose (25 g) and galactose (25 g); small intestine (without prior digestion) after
a normal rise in blood glucose indicates lactose ingestion (5 g dose) and excreted largely unchanged
intolerance. This test is less reliable than breath in urine. If excretion is less than 20% of ingested dose,
hydrogen test. absorption is considered as impaired (small intestinal
2. Breath hydrogen test: An oral dose of lactose is disease). Misleading results are obtained in renal
administered and breath hydrogen is measured. In failure, delayed gastric emptying and incomplete
lactase deficiency, lactose is not absorbed, and reaches urine collection.

Fig. 11.5: Diagnosis of lactose intolerance


Tests for Malabsorption and Pancreatic Function 131

Tests for bacterial overgrowth in small intestine Bacterial urine or of faecal water, while magnesium abuse can be
overgrowth in the small intestine occurs if there is stasis identified by measuring fecal osmotic gap and
of small bowel contents (e.g. due to stricture or magnesium concentration in fecal water.
diverticulosis). Bacteria cause deconjugation of bile salts
Other tests for malabsorption Some intestinal disorders
leading to failure of mixed micelle formation and fat
have characteristic radiological abnormalities like
malabsorption. Various tests for detection of small bowel
Crohn’s disease, diverticula, enterocolic fistula, etc. CT
bacterial overgrowth include:
scan can detect changes associated with chronic
• Aspiration and culture of duodenal/jejunal contents
pancreatitis.
• Breath hydrogen tests (e.g. with lactulose)
A definitive diagnosis can be established in certain
• Breath CO2 tests (e.g. with 14C-glycocholic acid or 14C-
malabsorption syndromes by endoscopically-directed
xylose)
biopsy of small intestinal mucosa.
• Measurement of urinary indican
1. Aspiration and culture of duodenal/jejunal contents:
PANCREATIC FUNCTION TESTS
Duodenal or jejunal contents are aspirated at
endoscopy and sent for bacterial culture. This is the The pancreas is a major gland having both exocrine and
most reliable diagnostic test for bacterial overgrowth; endocrine functions. The exocrine component secretes
however, it is invasive and occasionally false-negative its fluid into the pancreatic duct that opens into the
result is obtained. duodenum and is concerned with digestion of food. The
2. Breath hydrogen tests: Lactulose, a non-absorbable endocrine component secretes its hormones directly into
carbohydrate, is administered orally (10 g). In the bloodstream.
bacterial overgrowth, fermentation of unabsorbed
carbohydrate occurs in the small as well as large Anatomy and Physiology of Pancreas
intestine with the production of hydrogen. Breath 1. Exocrine component: Exocrine pancreas is the major
hydrogen is increased early (40 minutes) following component of the gland. It is made up of closely
the oral dose of lactulose. packed secretory acini that drain their secretions via
3. Breath CO 2 tests: In 14C-xylose breath test, 1 g small, highly branched ducts into the main pancreatic
radiolabeled xylose is given orally. In bacterial duct. The main pancreatic duct joins the common bile
overgrowth a large amount of xylose is metabolized duct and opens into the duodenum via the ampulla
by bacteria to 14CO2 that is absorbed and excreted of Vater. A small accessory pancreatic duct is also
through breath. This isotopically labeled CO 2 is present in most individuals and opens into the
measured in expired breath. duodenum separately (Fig. 11.6).
This test is better than 14C-glycocholic acid test. In
the later test, 14C-glycocholic acid (a radiolabeled
conjugated bile acid) is given orally. Normally this
bile acid is absorbed intact. Bacterial overgrowth
causes deconjugation of glycocholic acid with the
release of 14C-glycine which is absorbed in the small
intestine, and metabolized in the liver releasing
labeled carbon dioxide. Excretion of labeled CO2 is
measured in expired breath.
4. Test for urinary indican: Indican is a bacterial
metabolic product of dietary tryptophan.
Tests for laxative abuse Diarrhea due to malabsorption
should be distinguished from that due to laxative abuse.
Laxatives or their metabolites can be detected in urine
or loose fecal sample by thin layer chromatography.
Phenolphthalein can also be detected by alkalization of Fig. 11.6: Gross anatomy of pancreas
132 Essentials of Clinical Pathology

Each acinus is lined by pyramidal secretory cells, duodenum stimulates secretion of secretin that in turn
apices of which are packed with zymogen secretory stimulates release of bicarbonate from the pancreas into
granules (Figs 11.7 and 11.8). These granules contain the small intestine.
inactive forms of enzymes synthesized by acinar cells. Proteolytic enzymes are secreted in an inactive form
Zymogen granules are released into the acinar lumina (trypsinogen and chymotrypsinogen) into the pancreatic
by the process of exocytosis. Secretions in acinar lumina juice to prevent the autodigestion of pancreas. Entero-
initially drain into intercalated ducts, lining cells of which kinase, secreted by duodenal mucosa, activates the
secrete water and bicarbonate ions. High concentration inactive trypsinogen to trypsin in duodenal lumen.
of bicarbonate ions is responsible for alkaline pH of Trypsin converts chymotrypsinogen to the active enzyme
pancreatic juice, which serves to neutralize the acidic pH chymotrypsin. Trypsin and chymotrypsin degrade large
of the chyme entering into the duodenum. polypeptide molecules to smaller ones. Peptidase, an
The pancreatic enzymes entering into the duodenum intestinal enzyme bound to enterocyte cell membrane,
breakdown proteins, carbohydrates, lipids, and nucleic further degrades smaller polypeptides to amino acids.
acids, making them suitable for digestion and absorption. Other proteolytic enzymes elaborated by the pancreas
Pancreatic secretions also neutralize gastric acid entering include elastase, carboxypeptidases, ribonuclease, and
into the small intestine. Gastric acid entering the collagenase (Box 11.1).
Carbohydrates are broken down by amylase in the
pancreatic juice and by intestinal cell membrane-bound
disaccharidases to monosaccharides.
Lipids are emulsified in the duodenum by the action
of bile; they are then broken down by the pancreatic
lipase into free fatty acids, monoglycerides, and glycerol.
Exocrine pancreatic function is regulated by neural
factors and by certain hormones of the gastrointestinal
tract like cholecystokinin-pancreozymin (CCK-PZ),
gastrin, and secretin.
CCK-PZ stimulates contraction of gallbladder and
also secretion of enzyme-rich pancreatic juice. It is a
polypeptide hormone synthesized by duodenal endo-
crine cells in response to contact with partially digested
Fig. 11.7: Major components of exocrine pancreas proteins and gastric hydrochloric acid.
Gastrin is produced mainly by G cells of the antral
mucosa of the stomach and has actions on the pancreas
similar to that of CCK-PZ. Gastrin secretion is promoted

Box 11.1: Composition of pancreatic juice


• Digestive enzymes (secreted by exocrine acinar cells
of pancreas)
– Proteases: Digest proteins. Pancreatic proteases are
chymotrypsin and trypsin, which digest proteins to smaller
peptides. Other proteases are elastase, carboxypeptidase,
ribonuclease, and collagenase.
– Lipase: Digestion of dietary triglycerides to mono-
glycerides and free fatty acids
– Amylase: Hydrolysis of starch to maltose (a glucose-
glucose disaccharide)
• Bicarbonate and water (secreted by lining cells of
small pancreatic ducts): Neutralization of gastric acid
Fig. 11.8: Diagrammatic representation of pancreatic
entering duodenum
acini and terminal ductule
Tests for Malabsorption and Pancreatic Function 133

by antral distention and the presence of partially digested mild pancreatic insufficiency (i.e. when imaging studies
proteins. are normal). However, no such non-invasive pancreatic
Secretin is a polypeptide synthesized by duodenal function test is currently available. Also, the results of
endocrine S cells in response to contact with gastric these tests in pancreatic insufficiency often overlap with
hydrochloric acid. It enhances secretion of bicarbonate- normal values.
rich pancreatic fluid into the duodenum.
Normal pancreatic juice is colorless, has alkaline pH, Direct (Invasive or Tube) Tests
and its 24-hour volume is 1000-2500 ml. It consists of In direct tests, duodenum is intubated, a pancreatic
water, sodium, potassium, chloride, bicarbonate, and stimulant (such as secretin-CCK-PZ, secretin-cerulin, or
pancreatic enzymes. Lundh meal) is administered, and pancreatic secretions
2. Endocrine component: Isolated clusters of endocrine entering into the duodenum are aspirated. Quantity of
cells are scattered throughout the exocrine pancreatic bicarbonate and enzyme secretions is measured, which
tissue and are known as islets of Langerhans. correlates with functional mass of pancreas.
Endocrine cells are also present singly in between the 1. Secretin-CCK-PZ or Secretin-Cerulin test: This test
pancreatic acini. Hormones secreted by the endocrine is the ‘gold standard’ for detection of exocrine pancreatic
pancreas include insulin, glucagon, somatostatin, insufficiency.
vasoactive intestinal polypeptide, and pancreatic This test is based on the principle that output of
polypeptide. pancreatic juice, bicarbonate, and enzymes from pancreas
(after maximum stimulation by an exogenously adminis-
Tests of Pancreatic Function tered hormone) is dependent on the amount of functional
pancreatic tissue. The method, in short, is as follows:
Tests for assessing pancreatic function measure the
i. In a fasting patient, duodenum is intubated with a
physiologic function of exocrine component. These tests
radioopaque tube under fluoroscopic guidance and
either determine the functional activity of certain
contents are aspirated until the contents are clear.
pancreatic enzymes, or directly quantitate the products ii. Secretin and cholecystokinin-pancreozymin (or
of pancreatic secretions. secretin-caerulin) are administered intravenously.
Pancreatic function tests are listed in Table 11.2. Secretin stimulates secretion of watery, bicarbonate-
Nowadays, the direct tests have largely been replaced rich fluid, while CCK-PZ or cerulin stimulate
by the indirect (tubeless) tests. secretion of enzymes from the pancreas.
Pancreatic insufficiency can be readily diagnosed in iii. Three samples of duodenal secretions are aspirated:
advanced cases showing steatorrhea, pancreatic basal, following intravenous secretin, and following
calcification, and diabetes mellitus. Clinical features of intravenous CCK-PZ. Total volume of fluid, pH and
pancreatic insufficiency do not appear until about 90% concentrations of bicarbonate and enzymes (amylase,
of the gland is destroyed. If pancreatic insufficiency is trypsin) are measured, and compared with the
detected early, appropriate treatment can be given and normal values.
improved outlook can be expected. Ideally, exocrine In exocrine pancreatic insufficiency, bicarbonate
pancreatic function tests should be simple to perform, secretion is lost earlier than enzyme secretions. Also,
specific, sensitive, and should be able to detect early or enzyme activity and bicarbonate content are reduced
before there is any reduction in volume of pancreatic
juice.
Table 11.2: Pancreatic function tests
The specificity and sensitivity of this test is high
Direct tests (Invasive) (around 90%). However, the test is invasive, laborious,
• Secretin-CCK-PZ test (or secretin-cerulin test)
time-consuming, and expensive. It also requires special
• Lundh test
skills for performance and interpretation.
Indirect tests (Non-invasive) 2. Lundh meal: Lundh meal is a physiologic test meal
• NBT-PABA (Bentiromide) test consisting of protein (15 gm), fat (18 gm), carbohydrates
• Pancreolauryl test (40 gm glucose), and water (300 ml). It induces pancreatic
• Breath tests secretion by stimulating release of endogenous secretin
• Estimation of faecal enzymes
• Estimation of faecal fat and CCK-PZ. Following administration of the meal,
duodenal contents are aspirated and activity of one or
134 Essentials of Clinical Pathology

more enzymes (usually trypsin) and the volume of the ester hydrolase in the presence of bile acids to release
pancreatic juice are measured. The results are usually fluorescein, which is then absorbed in the small intestine,
abnormal in pancreatic insufficiency. As compared to the partially conjugated by the liver, and excreted in urine.
direct hormonal stimulation, this test is more physio- Amount of fluorescein excreted in urine for 10 hours is
logical and cheaper. measured (Alternatively, serum concentrations can also
Disadvantages of this test are: be measured). Test is repeated a day later, but following
i. Lower specificity and sensitivity (70-80%) as ingestion of free fluorescein to correct for individual
compared to the direct secretin-pancreozymin test. variation in intestinal absorption, conjugation in liver,
ii. Abnormal result is also obtained in small bowel and urinary excretion. Ratio of excretion of fluorescein
mucosal disease. dilaurate to excretion of free fluorescein is determined.
If it is less than 0.20, pancreatic insufficiency is present.
Indirect (Non-invasive or Tubeless) Tests
Sensitivity and specificity are similar to NBT-PABA test.
As direct tests are invasive, labor-intensive, and need a 3. Breath tests: Breath tests have been developed which
specialized set-up, various indirect (tubeless) function indirectly assess pancreatic function by measuring 13CO2
tests have been developed. Deficient activity of pancreatic or 14CO2 in breath following ingestion of a substrate such
enzymes is indirectly demonstrated by reduced digestion as 14C-triglyceride, 14C-triolein, mixed triglyceride, 13C-
or increased faecal excretion of fat, low concentration of D-hiolein, or cholesteryl octanoate.
pancreatic enzymes in feces or in blood, or decreased Principle of one such test using 14C-triglyceride is as
hydrolysis of ingested synthetic compounds causing their follows. When 14C-triglyceride is ingested, it is hydro-
reduced urinary excretion. Although simpler, indirect lysed by pancreatic lipase to free fatty acids and
tests have low sensitivity and specificity and cannot monoglycerides. After absorption, metabolism of free
detect mild pancreatic insufficiency. fatty acids ultimately leads to the formation of 14CO2 that
1. NBT-PABA test (Bentiromide test): When a synthetic is excreted in the breath. The amount of 14CO2 formed is
compound N-benzoyl-l tyrosyl-p-aminobenzoic acid proportional to the rate of absorption of fatty acids in
(NBT-PABA, bentiromide) is ingested orally, it is the intestine. Normally there is a peak radioactivity in
hydrolyzed by pancreatic chymotrypsin in duodenum breath after about 6 hours. Low radioactivity in breath
to release p-aminobenzoic acid (PABA). PABA is can be due to deficient fat digestion or deficient fat
absorbed in the small intestine and metabolized in the absorption. To distinguish between the two, the test is
liver. The urinary excretion of metabolites of PABA is repeated after 2 weeks using radiolabeled free fatty acids
measured for 6 hours; if excretion is less than 50% of (instead of radiolabeled triglycerides). If the result is
ingested dose, exocrine pancreatic insufficiency is normal, pancreatic insufficiency is present.
present. Thus, amount of PABA excreted in urine
indirectly reflects activity of pancreatic chymotrypsin and Disadvantages of the test are:
pancreatic function. • Time consuming.
False results are obtained in small bowel disease • Exposure to radioisotopes.
(decreased absorption), liver disease (defective conju- • Inability to detect mild pancreatic insufficiency.
gation), and renal disease (decreased excretion). To guard • False-positive test in respiratory or metabolic disease
against false results, urinary excretion of PABA after in which excretion of CO2 is affected.
administration of bentiromide is compared against 4. Estimation of fecal enzymes: Concentration of
urinary excretion of free radiolabeled PABA given at the enzymes secreted by the pancreatic acinar cells into the
same time. gut is lowered in pancreatic insufficiency and obstruction
2. Pancreolauryl test (Test for pancreatic arylesterase): of pancreatic duct. Output of elastase-1 or chymotrypsin
This test is similar in principle to NBT-PABA test. (both of which remain intact during their passage
Fluorescein dilaurate (Pancreolauryl), a synthetic ester, through the gut) is measured in a fecal sample, which
is ingested along with a standard breakfast. Fluorescein correlates well with their secretion from the pancreas into
dilaurate is hydrolysed by pancreas-specific cholesterol the duodenum.
Tests for Malabsorption and Pancreatic Function 135

Activity of elastase-1 is measured in a random fecal Elevation of serum amylase occurs in:
sample by immunoassay. Its concentration is reduced in • Pancreatic diseases: acute and chronic pancreatitis,
pancreatic insufficiency. The test is sensitive and specific. pseudocyst
Although the test is simple and noninvasive, it is • Parotitis
expensive. • Intestinal diseases: perforation, ischemia, obstruction.
Activity of chymotrypsin is measured by spectro- • Biliary tract disease
photometric assay of 4-nitroaniline in a fecal sample (A • Ectopic pregnancy
synthetic pentapeptide is mixed with fecal extract that is
• Malignant tumors of lung or ovary
hydrolyzed by chymotrypsin in feces to release 4-
• Macroamylasemia.
nitroaniline). Although the test is cheaper, it has low
sensitivity. It is commonly used as a screening test for Binding of normal serum amylase with high mole-
detection of pancreatic insufficiency in cystic fibrosis. cular weight plasma proteins (immunoglobulins) leads
5. Estimation of fecal fat: Quantitation of fecal fat is a to the formation of macroamylase. Because of large size,
definitive test for diagnosis of steatorrhea but not for they cannot be excreted in urine. Macroamylasemia is
identification of its cause. The standard indirect test for not associated with any clinical features, but it must be
pancreatic insufficiency is estimation of 72-hour faecal
distinguished from other causes of elevated serum
fat. (See Chapter 9: Examination of Feces).
amylase. In renal failure, and in macroamylasemia,
Normal fat excretion of fat is <7% of total amount of
fat ingested. In pancreatic insufficiency, fat excretion is urinary amylase is low or absent.
often >20%. 2. Serum lipase: Serum lipase is elevated in pancreatic
Aside from pancreatic insufficiency, excess fecal fat diseases, intestinal diseases, acute cholecystitis, and renal
excretion also occurs in conditions such as biliary failure. Values are normal in parotitis and in macro-
obstruction, small bowel mucosal disease, lymphatic amylasemia.
obstruction, liver disease, abetalipoproteinemia, and In acute pancreatitis, serum amylase begins to rise
small bowel bacterial overgrowth. Apart from its
within 3-6 hours, peaks at 24 hours, and returns to normal
nonspecificity, the test is inconvenient and unpleasant.
levels by 2-3 days. Urinary amylase is also high in acute
It cannot differentiate between malabsorption and
deficient digestion, and the result is normal in mild pancreatitis and remains elevated for 7-10 days. Serum
pancreatic insufficiency. lipase starts to increase within 3-6 hours, reaches
maximum at 24 hours, and remains elevated for 8-14
Pancreatic Enzymes Used as Markers of
days.
Active Pancreatic Disease
Very high levels of both serum amylase and serum
Two enzymes, serum or urinary amylase and serum
lipase (i.e. >5 times the upper limit of normal) are
lipase, are often measured to determine the presence of
observed in acute pancreatitis; in other intra-abdominal
pancreatic disease, especially acute pancreatitis.
1. Amylase: Serum amylase is mainly derived from disorders, elevations are moderate or slight. It has been
pancreas and salivary glands. Amylase is a small recommended to measure both serum amylase and
molecule and is filtered by the glomeruli and excreted in serum lipase if diagnosis of acute pancreatitis is suspected
urine. (Fig. 11.9).

Fig. 11.9: Evaluation of raised serum amylase


136 Essentials of Clinical Pathology

REFERENCE RANGES Urinary excretion of


fluorescein dilaurate
• Fecal fat (24 hour): < 7 g/24 hours • Pancreolauryl test: ——————————————>0.30
Urinary excretion of
• Fecal fat globules: Average 2.5 per high power field free dilaurate
in random sample
• 14CO breath test: Peak radioactivity at 5-7 hours
• Fecal elastase: >190 μg/g feces
2 • Fecal chymotrypsin: 120-1265 μg/g (Mean: 290 μg/g)
• D-xylose absorption test: >1.2 g/5 hour (5 g dose); • Serum amylase: 35-115 U/L
> 4.0 g/5 hours (25.0 g dose) • Serum lipase: 56-239 U/L
• Lactulose breath hydrogen test: 30-40 minutes
• Lactose tolerance test: Blood glucose >30 mg/dl above BIBLIOGRAPHY
fasting value.
1. Dimagno EP. A perspective on the use of tubeless
Urinary excretion of
pancreatic function tests. Gut 1998;43:2-3.
PABA
• NBT-PABA (Bentiromide) test:——————————> 0.75 2. Provan D and Krentz A. Oxford Handbook of Clinical
Urinary excretion of and Laboratory Investigation. Oxford University Press.
free PABA Oxford, 2002.
12

Thyroid Function Tests

ANATOMY AND PHYSIOLOGY


The thyroid gland is a butterfly-shaped endocrine gland
composed of two lateral lobes connected by a thin band
of tissue called as isthmus. It is located in the neck in
front of the upper part of trachea (Fig. 12.1). Normal
weight is 15 to 25 grams.
The thyroid follicle is the functional unit of the thyroid
gland. It consists of a single layer of cuboidal epithelial
cells that rest on a basement membrane and enclose
amorphous material called as colloid (Fig. 12.2). The
follicles synthesize and secrete iodine-containing
hormones tri-iodothyronine (T3) and tetra-iodothyronine
or thyroxine (T4). The colloid consists mainly of
thyroglobulin (Tg) that stores the thyroid hormones
before secretion. Fig. 12.1: Anatomy of thyroid gland
The thyroid gland also consists of a second type of
cell called as parafollicular or C (clear) cells that are
scattered amongst the follicular cells. These cells have a
completely distinct embryological origin, and synthesize
and secrete the hormone calcitonin, which regulates
blood calcium levels along with parathyroid hormone.
The thyroid gland plays a major role in the meta-
bolism and growth and development of tissues.
Biosynthesis of thyroid hormones: Iodine is essential for
synthesis of thyroid hormones. Iodine is obtained from
the diet; dietary content of iodine is dependent on
availability of iodine in land and water in a particular
region. Various steps in the synthesis of thyroid
hormones are as follows (Fig. 12.3):
• Trapping of inorganic iodide circulating in blood by
follicular cells by means of an iodide pump (sodium
iodide symporter)
• Diffusion of iodide towards the apical surface of Fig. 12.2: Normal histology of thyroid showing follicles
follicular cell lined by follicular cells and containing colloid
138 Essentials of Clinical Pathology

Fig. 12.3: Synthesis of thyroid hormones

• Oxidation of iodide to iodine by thyroid peroxidase


• Iodination of tyrosine residues of thyroglobulin
(organification) to form monoiodotyrosine (MIT) and
diiodotyrosine (DIT). This process occurs at the
interface of follicular cell and colloid. The rate of
synthesis of DIT is twice that of MIT.
Fig. 12.4: Regulation of thyroid hormone production.
• Coupling of MIT and DIT to form T3 (MIT+DIT) +: Stimulation; –: Inhibition
and T4 (DIT + DIT).
• Endocytosis of colloid droplets by follicular cell and
proteolysis of Tg-hormone complex leads to the
formation of a cytoplasmic vacuole within the cell.
Fusion of lysosome with the vacuole is followed by
action of hydrolytic enzymes and release of the
hormone from the thyroglobulin. MIT and DIT are
deiodinated for recycling of iodine.
• T3 and T4 are released into the circulation. Traces
of thyroglobulin are also released.
Thyroid stimulating hormone (TSH) from anterior
lobe of the pituitary gland regulates the synthesis and
secretion of thyroid hormones by a feedback mechanism.
Thyrotropin releasing hormone (TRH) produced by
hypothalamus acts on pituitary to stimulate synthesis
and release of TSH. An increase in the level of thyroid
hormone inhibits the release of TRH from hypothalamus
and TSH from pituitary, while fall in the level of thyroid
hormone leads to an increase in the level of TRH and
TSH (Fig. 12.4).

Metabolism of Thyroid Hormones


Thyroid gland primarily secretes free or unbound T4 in Fig. 12.5: Metabolism of thyroid hormones.
circulation. Only a small amount of T3 is secreted directly rT3: Reverse T3
Thyroid Function Tests 139

Fig. 12.6: In conditions with altered TBG concentration, total T4 is altered. Therefore measurement of free
T4 is more reliable as a test of thyroid function. TBG: Thyroid-binding globulin

by the thyroid gland (10%). Majority of T3 in circulation


Box 12.1: Terminology in thyroid disorders
(90%) is derived from removal of one iodothyronine unit
from T4 (Fig. 12.5). T4 may also be converted in liver to • Primary hyper-/hypothyroidism: Increased or decreased
reverse T3 (rT3) which is an inactive hormone. function of thyroid gland due to disease of thyroid itself
In circulation, T4 and T3 are bound reversibly to and not due to increased or decreased levels of TRH
or TSH.
carrier proteins like thyroxine-binding globulin (TBG),
• Secondary hyper-/hypothyroidism: Increased or
thyroxine-binding prealbumin (TBPA), and albumin.
decreased function of thyroid gland due to increased or
Very small amounts of T3 (0.5%) and T4 (0.05%) are decreased levels of TSH.
unbound and free. Only the free forms of the hormone • Tertiary hypothyroidism: Decreased function of thyroid
(T3 and T4) are biologically active. T3 is more potent gland due to decreased function of hypothalamus.
than T4, although levels of T4 are 50-times more than • Subclinical thyroid disease: A condition with abnormality
those of T3. Variations in the levels of carrier proteins of thyroid hormone levels in blood but without specific
affect the levels of total T4 (Fig. 12.6) and T3, even in clinical manifestations of thyroid disease and without any
history of thyroid dysfunction or therapy.
normal individuals e.g. increased TBG concentration
• Subclinical hyperthyroidism: A condition with normal
occurs during pregnancy due to stimulation of its thyroid hormone levels but with low or undetectable TSH
synthesis in liver by high oestrogen levels. Total T4 and level.
T3 levels are dependent on thyroid function and level of • Subclinical hypothyroidism: A condition with normal
TBG, and therefore total levels will be abnormal if TBG thyroxine and triiodothyronine level along with mildly
is abnormal Therefore, free levels of T4 and T3 are better elevated TSH level.
indicators of thyroid status than total levels.
Table 12.1: Causes of hyperthyroidism
DISORDERS OF THYROID 1. Graves’ disease (Diffuse toxic goiter)
Among the endocrine disorders, disorders of thyroid 2. Toxicity in multinodular goiter
are common and are only next in frequency to diabetes 3. Toxicity in adenoma
mellitus. They are more common in women than in men. 4. Subacute thyroiditis
Functional thyroid disorders can be divided into two 5. TSH-secreting pituitary adenoma (secondary hyperthy-
types depending on activity of the thyroid gland: roidism)
hypothyroidism (low thyroid hormones), and hyper- 6. Trophoblastic tumours that secrete TSH-like hormone:
choriocarcinoma, hydatidiform mole
thyroidism (excess thyroid hormones). Any enlargement
7. Factitious hyperthyroidism
of thyroid gland is called as a goiter. Terminology related
to thyroid disorders is shown in Box 12.1.
Clinical Characteristics
Hyperthyroidism Clinical characteristics of hyperthyroidism are nervous-
ness, anxiety, irritability, insomnia, fine tremors; weight
Hyperthyroidism is a condition caused by excessive loss despite normal or increased appetite; heat intole-
secretion of thyroid hormone. Causes of hyperthyroidism rance; increased sweating; dyspnea on exertion;
are listed in Table 12.1. amenorrhea and infertility; palpitations, tachycardia,
140 Essentials of Clinical Pathology

cardiac arrhythmias, heart failure (especially in elderly); Table 12.2: Differences between primary and
and muscle weakness, proximal myopathy, and secondary hyperthyroidism
osteoporosis (especially in elderly). Parameter Primary Secondary
The triad of Graves’ disease consists of hyper- hyperthyroidism hyperthyroidism
thyroidism, ophthalmopathy (exophthalmos, lid
1. Serum TSH Low Normal or high
retraction, lid lag, corneal ulceration, impaired eye
2. Serum free High High
muscle function), and dermopathy (pretibial myxo- thyroxine
edema). 3. TSH receptor May be Negative
antibodies positive
Laboratory Features 4. Causes Graves’ disease,
In most patients, free serum T3 and T4 are elevated. In toxic multinodular
goiter, toxic Pituitary
T3 thyrotoxicosis (5% cases of thyrotoxicosis), serum T4
adenoma adenoma
levels are normal while T3 is elevated. Serum TSH is low
or undetectable (< 0.1 mU/L) (Box 12.2).
Undetectable or low serum TSH along with normal Hypothyroidism
levels of T3 and T4 is called as subclinical hyper- Hypothyroidism is a condition caused by deficiency of
thyroidism; subtle signs and symptoms of thyrotoxicosis thyroid hormones. Causes of hypothyroidism are listed
may or may not be present. Subclinical hyperthyroidism in Table 12.3. Primary hypothyroidism results from
is associated with risk of atrial fibrillation, osteoporosis, deficient thyroid hormone biosynthesis that is not due
and progression to overt thyroid disease. to disorders of hypothalamus or pituitary. Secondary
Features of primary and secondary hyperthyroidism hypothyroidism results from deficient secretion of TSH
are compared in Table 12.2. from pituitary. Deficient or loss of secretion of thyro-
Evaluation of hyperthyroidism is presented in Figure
12.7.
Table 12.3: Causes of hypothyroidism

Box 12.2: Thyroid function tests in hyperthyroidism 1. Primary hypothyroidism (Increased TSH)
• Iodine deficiency
• Thyrotoxicosis: • Hashimoto’s thyroiditis
– Serum TSH low or undetectable • Exogenous goitrogens
– Raised total T4 and free T4. • Iatrogenic: surgery, drugs, radiation
• T3 toxicosis: 2. Secondary hypothyroidism (Low TSH): Diseases of
– Serum TSH undetectable pituitary
– Normal total T4 and free T4 3. Tertiary hypothyroidism (Low TSH, Low TRH):
– Raised T3 Diseases of hypothalamus

Fig. 12.7: Evaluation of hyperthyroidism. TSH: thyroid stimulating hormone; FT4: free T4; FT3: free T3; TRAb: TSH
receptor antibody; TRH: Thyrotropin releasing hormone
Thyroid Function Tests 141

Table 12.4: Differences between primary and secondary hypothyroidism

Parameter Primary Secondary


hypothyroidism hypothyroidism

1. Cause Hashimoto’s thyroiditis Pituitary disease


2. Serum TSH High Low
3. Thyrotropin releasing hormone Exaggerated response No response
stimulation test
4. Antimicrosomal antibodies Present Absent

tropin releasing hormone from hypothalamus results in


Box 12.3: Thyroid function tests in hypothyroidism
tertiary hypothyroidism. Secondary and tertiary
hypothyroidism are much less common than primary. • Primary hypothyroidism
Plasma TSH is high in primary and low in secondary – Serum TSH: Increased (proportional to degree of
and tertiary hypothyroidism. Differences between hypofunction)
primary and secondary hypothyroidism are shown in – Free T4: Decreased
Table 12.4. – TRH stimulation test: Exaggerated response
Clinical features of primary hypothyroidism are:
lethargy, mild depression, disturbances in menstruation, • Secondary hypothyroidism
weight gain, cold intolerance, dry skin, myopathy, – Serum TSH: Decreased
– Free T4: Decreased
constipation, and firm and lobulated thyroid gland (in
– TRH stimulation test: Absent response
Hashimoto’s thyroiditis).
In severe cases, myxoedema coma (an advanced stage • Tertiary hypothyroidism
with stupor, hypoventilation, and hypothermia) can – Serum TSH: Decreased
occur. – FT4: Decreased
– TRH stimulation test: Delayed response
Laboratory Features
Laboratory features in hypothyroidism are shown in Box
12.3. obstetrical outcome, poor cognitive development in
Normal serum thyroxine (T4 and FT4) coupled with children, and high risk of hypercholesterolemia and
a moderately raised TSH (>10 mU/L) is referred to as progression to overt hypothyroidism.
subclinical hypothyroidism. It is associated with bad Evaluation of hypothyroidism is presented in Figure 12.8.

Fig. 12.8: Evaluation of hypothyroidism. TSH: thyroid stimulating hormone;


FT4: free T4; TRH: Thyrotropin releasing hormone
142 Essentials of Clinical Pathology

THYROID FUNCTION TESTS Table 12.5: Causes of low and increased TSH

Biochemical tests for diagnosis of a thyroid disorder are Low TSH Increased TSH
called as thyroid function tests. The first-line tests are
Primary hyperthyroidism Primary hypothyroidism
serum TSH, total T4 or free T4, and total T3 and free T3.
A variety of non-thyroidal diseases can alter the T3 toxicosis Secondary hyperthyroidism
(pituitary adenoma secreting
results of thyroid function tests in patients with normal TSH)
thyroid status. These disorders include infections, liver
Secondary and tertiary
disease, malignancy, trauma, surgery, renal failure, and hypothyroidism
cardiac failure. To avoid misinterpretation, thyroid
function tests should not be performed during an acute
non-thyroidal illness. 2. Screening for hypothyroidism in newborn
3. Diagnosis of primary and secondary hypothyroidism,
Thyroid Stimulating Hormone (TSH)
and subclinical hypothyroidism
Currently, the most important single test to assess 4. Diagnosis of clinical and subclinical hyperthyroidism
thyroid function and to monitor thyroid hormone 5. Follow-up of T3 and T4 replacement therapy in
replacement therapy is a sensitive TSH assay. This is hypothyroidism.
because most cases of thyroid dysfunction result from
primary thyroid disease. TSH is a hormone secreted by The best single test for initial assessment of thyroid
anterior pituitary gland. A normal TSH level excludes function is a sensitive (3rd or 4th generations) TSH
thyroid disease. TSH is low in primary hyperthyroidism assay, which is sufficiently sensitive and specific for
and high in primary hypothyroidism. The standard early detection of primary hyper- and hypothyroidism.
method for measurement of TSH is immunoassay. Newer
• In primary hyperfunctioning of thyroid, TSH is low.
sensitive methods can reliably distinguish between
• In primary hypofunction of thyroid, TSH is high.
extremely low or undetectable levels seen in hyper-
• Serum free T4 should be measured in all patients
thyroidism and low normal or below-normal levels seen
with elevated TSH.
in some euthyroid patients.
• Serum free T4 and total T3 or free T3 should be
Normal reference range in adults is 0.5 –5.0 mU/L
measured in all patients with low TSH.
and in newborns < 20 mU/L. In adults, borderline
increase is 5-10 mU/L, while values >10 mU/L are
considered as high. Values less than 0.1 mU/L are low. TOTAL THYROXINE (T4)
Third and fourth generations TSH assays have detection
Total serum thyroxine includes both free and protein-
limits of 0.01 to 0.02 mU/L and 0.001 to 0.002 mU/L,
bound thyroxine and is usually measured by competitive
respectively.
immunoassay. Normal level in adults is 5.0-12.0 μg/dl.
TSH levels are affected by non-thyroidal illness and
Test for total thyroxine or free thyroxine is usually
certain drugs; therefore for confirmation of thyroid
combined with TSH measurement and together they
dysfunction, estimation of free T4 and total T4 should
give the best assessment of thyroid function.
also be done. With the availability of sensitive TSH
assays, TRH stimulation test is usually not required (see
Causes of Increased Total T4
later).
Causes of low and increased TSH are shown in Table 1. Hyperthyroidism: Elevation of both T4 and T3 values
12.5. along with decrease of TSH are indicative of primary
hyperthyroidism.
Uses of TSH Measurement
2. Increased thyroxine-binding globulin: If concentration
1. Screening for euthyroidism: The availability of of TBG increases, free hormone level falls, release of
sensitive TSH assay has made the TSH measurement TSH from pituitary is stimulated, and free hormone
the first-line test for assessment of thyroid function. concentration is restored to normal. Reverse occurs
In most patients a normal TSH test indicates normal if concentration of binding proteins falls. In either
function, and no further testing is required case, level of free hormones remains normal, while
Thyroid Function Tests 143

concentration of total hormone is altered. Therefore, Free T3: Measurement of free T3 gives true values in
estimation of only total T4 concentration can cause patients with altered serum protein levels (like preg-
misinterpretation of results in situations that alter nancy, intake of estrogens or oral contraceptives, and
concentration of TBG. nephrotic syndrome). It represents 0.5% of total T3.
3. Factitious hyperthyroidism
4. Pituitary TSH-secreting tumor. Thyrotropin Releasing Hormone (TRH)
Stimulation Test
Causes of Decreased Total T4
Uses
1. Primary hypothyroidism: The combination of
decreased T4 and elevated TSH are indicative of 1. Confirmation of diagnosis of secondary hypo-
primary hypothyroidism. thyroidism
2. Secondary or pituitary hypothyroidism 2. Evaluation of suspected hypothalamic disease
3. Tertiary or hypothalamic hypothyroidism 3. Suspected hyperthyroidism
4. Hypoproteinaemia, e.g. nephrotic syndrome This test is not much used nowadays due to the
5. Drugs: oestrogen, danazol availability of sensitive TSH assays.
6. Severe non-thyroidal illness.
Procedure
Free Thyroxine (FT4)
• A baseline blood sample is collected for estimation
FT4 comprises of only a small fraction of total T4, is
of basal serum TSH level.
unbound to proteins, and is the metabolically active form
• TRH is injected intravenously (200 or 500 μg) followed
of the hormone. It constitutes about 0.05% of total T4.
by measurement of serum TSH at 20 and 60 minutes.
Normal range is 0.7 to 1.9 ng/dl. Free hormone
concentrations (FT4 and FT3) correlate better with
Interpretation
metabolic state than total hormone levels (since they are
not affected by changes in TBG concentrations). . 1. Normal response: A rise of TSH > 2 mU/L at 20
Measurement of FT4 is helpful in those situations in minutes, and a small decline at 60 minutes.
which total T4 level is likely to be altered due to alteration 2. Exaggerated response: A further significant rise in
in TBG level (e.g. pregnancy, oral contraceptives, already elevated TSH level at 20 minutes followed
nephrotic syndrome). by a slight decrease at 60 minutes; occurs in primary
hypothyroidism.
Total and Free Triiodothyronine (T3) 3. Flat response: There is no response; occurs in
Uses secondary (pituitary) hypothyroidism.
4. Delayed response: TSH is higher at 60 minutes as
1. Diagnosis of T3 thyrotoxicosis: Hyperthyroidism with compared to its level at 20 minutes; seen in tertiary
low TSH and elevated T3, and normal T4/FT4 is
(hypothalamic) hypothyroidism.
termed T3 thyrotoxicosis.
2. Early diagnosis of hyperthyroidism: In early stage of Antithyroid Antibodies
hyperthyroidism, total T4 and free T4 levels are
normal, but T3 is elevated. Various autoantibodies (TSH receptor, antimicrosomal,
A low T3 level is not useful for diagnosis of and antithyroglobulin) are detected in thyroid disorders
hypothyroidism since it is observed in about 25% of like Hashimoto’s thyroiditis and Graves’ disease.
normal individuals. Antimicrosomal (also called as thyroid peroxidase) and
For routine assessment of thyroid function, TSH and anti-thyroglobulin antibodies are observed in almost all
T4 are measured. T3 is not routinely estimated because patients with Hashimoto’s disease. TSH receptor
normal plasma levels are very low. antibodies (TRAb) are mainly tested in Graves’ disease
Normal T3 level is 80-180 ng/dl. to predict the outcome after treatment (Box 12.4).
144 Essentials of Clinical Pathology

– Toxic multinodular goiter: Multiple discrete areas


Box 12.4: Thyroid autoantibodies
of increased uptake
• Useful for diagnosis and monitoring of autoimmune thyroid – Adenoma: Single area of increased uptake
diseases. • Evaluation of a solitary thyroid nodule:
• Antimicrosomal or antithyroid peroxidase antibodies: – ‘Hot’ nodule: Hyperfunctioning
Hashimoto’s thyroiditis – ‘Cold’ nodule: Non-functioning; about 20% cases
• Anti-TSH receptor antibodies: Graves’ disease are malignant.
Interpretation of thyroid function tests is shown in
Table 12.6.
Radioactive Iodine Uptake (RAIU) Test
This is a direct test that assesses the trapping of iodide Table 12.6: Interpretation of thyroid function tests
by thyroid gland (through the iodine symporters or Test results Interpretations
pumps in follicular cells) for thyroid hormone synthesis.
Patient is administered a tracer dose of radioactive iodine 1. TSH Normal, FT4 Normal Euthyroid
2. Low TSH, Low FT4 Secondary hypothyroidism
(131I or 123I) orally. This is followed by measurement of
3. High TSH, Normal FT4 Subclinical hypothyroidism
amount of radioactivity over the thyroid gland at 2 to 6
4. High TSH, Low FT4 Primary hypothyroidism
hours and again at 24 hours. RAIU correlates directly
5. Low TSH, Normal FT4, Subclinical
with the functional activity of the thyroid gland. Normal Normal FT3 hyperthyroidism
RAIU is about 10-30% of administered dose at 24 hours, 6. Low TSH, Normal FT4, T3 toxicosis
but varies according to the geographic location due to High FT3
differences in dietary intake. 7. Low TSH, High FT4 Primary hyperthyroidism

Causes of Increased Uptake Neonatal Screening for Hypothyroidism

• Hyperthyroidism due to Graves’ disease, toxic Thyroid hormone deficiency during neonatal period can
multinodular goiter, toxic adenoma, TSH-secreting cause severe mental retardation (cretinism) that can be
tumor. prevented by early detection and treatment. Estimation
of TSH is done on dry blood spots on filter paper or cord
Causes of Decreased Uptake serum between 3rd to 5th days of life. Elevated TSH is
diagnostic of hypothyroidism. In infants with confirmed
• Hyperthyroidism due to administration of thyroid hypothyroidism, RAIU (123I) scan should be done to
hormone, factitious hyperthyroidism, subacute distinguish between thyroid agenesis and dyshormono-
thyroiditis. genesis.

Uses REFERENCE RANGES


RAIU is most helpful in differential diagnosis of 1. Total serum thyroxine (T4): 5.0-12.0 μg/dl
hyperthyroidism by separating causes into those due to 2. Free thyroxine (FT4): 0.7-1.9 ng/dl
increased uptake and due to decreased uptake. 3. Free thyroxine index (FTI): 4-11
4. Total tri-iodothyronine (T3): 80-180 ng/dl
Thyroid Scintiscanning 5. Free tri-iodothyronine (FT3): 2.3-4.2 pg/ml
5. Radioactive iodine uptake: 10-30%
An isotope (99mTc-pertechnetate) is administered and a
6. Thyroid stimulating hormone: 0.5-5.0 mU/L
gamma counter assesses its distribution within the
7. Serum reverse T3 (rT3): 10-40 ng/dl
thyroid gland.
Interpretation BIBLIOGRAPHY
• Differential diagnosis of high RAIU thyrotoxicosis:
– Graves’ disease: Uniform or diffuse increase in 1. Demers LM. Thyroid disease: pathophysiology and
uptake diagnosis. Clin Lab Med 2004;24:19-28.
Thyroid Function Tests 145

2. Heuck CC, Kallner A, Kanagasabapathy AS, Riesen W. 5. McDermott MT. Endocrine Secrets (4th Ed).
Diagnosis and monitoring of diseases of thyroid. World Philadelphia. Mosby, 2005.
Health Organization. 2000 WHO/DIL/0.004. 6. US Preventive Services Task Force: Screening for
3. Kaplan MM. Clinical perspectives in the diagnosis of thyroid disease: Recommended statement. Ann Intern
thyroid disease. Clin Chem 1999;45:1377-83. Med 2004;140:125-7.
4. Lazarus JH, Obuobie K. Thyroid disorders—an update. 7. Woeber KA. The year in review: the thyroid. Ann Intern
Postgrad Med J 2000;76:529-36. Med 1999;131:959-62.
13

Pregnancy Tests

Pregnancy tests detect human chorionic gonadotropin CLINICAL APPLICATIONS OF TESTS FOR
(hCG) in serum or urine. Although pregnancy is the most HUMAN CHORIONIC GONADOTROPIN
common reason for ordering the test for hCG, measure-
1. Early diagnosis of pregnancy: Qualitative serum hCG
ment of hCG is also indicated in other conditions as
test becomes positive 3 weeks after last menstrual
shown in Box 13.1.
period (LMP), while urine hCG test becomes positive
Human chorionic gonadotropin is a glycoprotein
5 weeks after LMP.
hormone produced by placenta that circulates in
2. Exclusion of pregnancy before prescribing certain
maternal blood and excreted intact by the kidneys. It
medications (like oral contraceptives, steroids, some
consists of two polypeptide subunits: α (92 amino acids)
antibiotics), and before ordering radiological studies,
and β (145 amino acids) which are non-covalently bound
radiotherapy, or chemotherapy. This is necessary to
to each other. Structurally, hCG is closely related to three
prevent any teratogenic effect on the fetus.
other glycoprotein hormones, namely, luteinizing
3. Early diagnosis of ectopic pregnancy: Trans-vaginal
hormone (LH), follicle-stimulating hormone (FSH), and
ultrasonography (USG) and quantitative estimation
thyroid-stimulating hormone (TSH). The α subunits of
of hCG are helpful in early diagnosis of ectopic
hCG, LH, FSH, and TSH are similar, while β subunits
pregnancy (before rupture).
differ and confer specific biologic and immunologic
4. Evaluation of threatened abortion: Serial quantitative
properties. Immunological tests use antibodies directed
estimation of hCG is helpful in following the course
against β-subunit of hCG to avoid cross-reactivity against
of threatened abortion.
LH, FSH, and TSH.
5. Diagnosis and follow-up of gestational trophoblastic
Syncytiotrophoblastic cells of conceptus and later of
disease (GTD).
placenta synthesize hCG. Human chorionic gonado-
6. Maternal triple test screen: This consists of measure-
tropin supports the corpus luteum of ovary during early
ment of hCG, α-fetoprotein, and unconjugated estriol
pregnancy. Progesterone, produced by corpus luteum,
in maternal serum at 14-19 weeks of gestation. The
prevents ovulation and thus maintains pregnancy. After
maternal triple screen identifies pregnant women
7-10 weeks of gestation, sufficient amounts of proges-
with increased risk of Down syndrome and major
terone are synthesized by placenta, and hCG is no longer
congenital anomalies like neural tube defects.
needed and its level declines.
7. Follow-up of ovarian or testicular germ cell tumors,
which produce hCG.
Box 13.1: Indications for measurement of β human
chorionic gonadotropin Normal Pregnancy
• Early diagnosis of pregnancy In women with normal menstrual cycle, conception
• Diagnosis and management of gestational trophoblastic (fertilization of ovum to form a zygote) occurs on day 14
disease in the fallopian tube. Zygote travels down the fallopian
• As a part of maternal triple test screen
tube into the uterus. Division of zygote produces a
• Follow-up of malignant tumors that produce β human
chorionic gonadotropin. morula. At 50-60-cell stage, morula develops a primitive
yolk sac and is then called as a blastocyst. About 5 days
Pregnancy Tests 147

after fertilization, implantation of blastocyst occurs in the


Box 13.2: Diagnosis of early pregnancy
uterine wall. Trophoblastic cells (on the outer surface of
the blastocyst) penetrate the endometrium and develop • Positive serum hCG test: 8 days after conception or 3
into chorionic villi. There are two main forms of weeks after last menstrual period (LMP)
• Positive urine hCG test: 21 days after conception or 5
trophoblasts—syncytiotrophoblast and cytotrophoblast.
weeks after LMP
Placental development occurs from chorionic villi. After • Ultrasonography for visualization of gestational sac:
formation of placenta, the conceptus is called as an – Transvaginal: 21 days after conception or 5 weeks
embryo. When embryo develops most major organs, it after LMP
is called as fetus (after 10 weeks of gestation). – Transabdominal: 28 days after conception or 6 weeks
after LMP
Human chorionic gonadotropin is synthesized by
syncytiotrophoblasts (of placenta) and detectable
amounts (~5 mIU/ml) appear in maternal serum about evaluation of ectopic pregnancy, failing pregnancy,
8 days after conception (3 weeks after LMP). In the first and for follow-up of gestational trophoblastic disease.
trimester (first 12 weeks, calculated from day 1 of LMP)
of pregnancy, hCG levels rapidly rise with a doubling Ectopic Pregnancy
time of about 2 days. Highest or peak level is reached at Ectopic pregnancy refers to the implantation of blastocyst
8-10 weeks (about 100,000 mIU/ml). This is followed by at a site other than the cavity of uterus. The most common
a gradual fall, and from 15-16 weeks onwards, a steady of such sites (>95% cases) is fallopian tube. Early diagnosis
level of 10,000-20,000 mIU/ml is maintained for the rest and treatment of tubal ectopic pregnancy is essential since
of the pregnancy (Fig. 13.1). After delivery, hCG becomes it can lead to maternal mortality (from rupture and
non-detectable by about 2 weeks. hemorrhage) and future infertility. Ectopic pregnancy is
Box 13.2 shows minimum time required for the a leading cause of maternal death during first trimester.
earliest diagnosis of pregnancy by hCG test and Diagnosis of ectopic pregnancy can be readily made in
ultrasonography (USG). most cases by ultrasonography and estimation of β-
subunit of human chorionic gonadotropin.
Two types of pregnancy tests are available: Early diagnosis of unruptured tubal pregnancy can
• Qualitative tests: These are positive/negative result be made by quantitative estimation of serum hCG and
types that are done on urine sample. ultrasonography. In normal intrauterine pregnancy, hCG
• Quantitative tests: These give numerical result and titer doubles every 2 days until first 40 days of gestation.
are done on serum or urine. They are also used for If hCG rise is abnormally slow, then an unviable
pregnancy (either ectopic or abnormal intrauterine
pregnancy) should be suspected.
Transabdominal USG can detect gestational sac in
intrauterine pregnancy 6 weeks after LMP. The level of
hCG in serum at this stage is >6500 mIU/ml. If
gestational sac is not visualized at this level of hCG, then
there is a possibility of ectopic pregnancy. Transvaginal
ultrasonography can detect ectopic pregnancy average
1 week earlier than abdominal ultrasonography; it can
detect gestational sac if β-hCG level is 1000-1500 mIU/
ml. Therefore, if gestational sac is not visualized in the
presence of >1500 mIU/ml of β-hCG level, an ectopic
pregnancy can be suspected.
Early diagnosis of ectopic pregnancy provides the
option of administration of intramuscular methotrexate
(rather than surgery), which causes dissolution of
Fig. 13.1: Level of human chorionic gonadotropin conceptus. This improves the chances of patient’s future
during pregnancy fertility.
148 Essentials of Clinical Pathology

Serial measurements of hCG after surgical removal LABORATORY TESTS FOR


of ectopic pregnancy can help in detecting persistence HUMAN CHORIONIC GONADOTROPIN
of trophoblastic tissue.
These are classified into two main groups:
• Biological assays or bioassays
Abortion
• Immunological assays
Termination of pregnancy before fetus becomes viable
(i.e. before 20 weeks) is called as abortion. Bioassays
In threatened abortion, vaginal bleeding is present In bioassay, effect of hCG is tested on laboratory animals
but internal os is closed and process of abortion, though under standardized conditions. There are several
started, is still reversible. It is possible that pregnancy limitations of bioassays like need for animal facilities,
will continue. need for standardization of animals, long time required
Serial quantitative titers of hCG showing lack of for the test results, low sensitivity, and high cost.
expected doubling of hCG level and USG are helpful in Therefore, bioassays have been replaced by immuno-
diagnosis and management of abortion. logical assays.
In Ascheim-Zondek test, urine from pregnant woman
Gestational Trophoblastic Disease (GTD) is injected into immature female mice. Formation of
It is characterized by proliferation of pregnancy- hemorrhagic corpora lutea in ovaries (after 4 days) is a
positive test. Friedman test is similar except that urine is
associated trophoblastic tissue. The two main forms of
injected into female rabbit. In rapid rat test, injection of
GTD are hydatidiform (vesicular) mole (benign) and
urine containing hCG into female rats is followed by
choriocarcinoma (malignant). Clinical features of GTD
hyperaemia and hemorrhage in ovaries. Yet another test
are as follows:
measures release of spermatozoa from male frog after
• Short history of amenorrhea followed by vaginal
injection of urine containing hCG.
bleeding.
• Size of uterus larger than gestational age; uterus is
Immunological Assays
soft and doughy on palpation with no fetal parts and
no fetal heart sounds. These are rapid and sensitive tests for detection and
• Excessive nausea and vomiting due to high hCG. quantitation of hCG. Variable results are obtained by
• Characteristic snowstorm appearance on pelvic USG. different immunological tests with the same serum
sample; this is due to differences in specificity of different
Quantitative estimation of hCG is helpful in diagnosis
immunoassays to complete hCG, β-subunit, and β-core
and management of GTD.
fragment. A number of immunological tests are
Trophoblastic cells of GTD produce more hCG as commercially available based on different principles like
compared to the trophoblasts of normal pregnancy for agglutination inhibition assay, enzyme immunoassay
the same gestational age. Concentration of hCG parallels including enzyme linked immunosorbent assay or
tumor load. Also, hCG continues to rise beyond 10 weeks ELISA, radioimmunoassay (RIA), and immunoradio-
of gestation without reaching plateau (as expected at the metric assay.
end of first trimester). A commonly used qualitative urine test is aggluti-
After evacuation of uterus, weekly estimation of hCG nation inhibition assay. Early morning urine specimen
is preferred because it contains the highest concentration
is advised till subsequent three (weekly) results are
of hCG. Causes of false-positive test include red cells,
negative; following evacuation of vesicular mole, hCG
leukocytes, bacteria, some drugs, proteins, and excess
becomes undetectable (after 2-3 months) on follow-up luteinizing hormone (menopause, midcycle LH surge)
in 80% of cases. Plateau or rising hCG indicates persistent in urine. Some patients have anti-mouse antibodies (that
GTD. In such cases, chemotherapy is indicated. are used in the test), while others have hCG-like material
Negative results for hCG after therapy should be in circulation, producing false-positive test. Anti-mouse
regularly followed up every 3 months for 1-2 years. antibodies also interfere with other antibody-based tests
Pregnancy Tests 149

hCG serum is neutralized, and no agglutination of latex


particles occurs (positive test). If there is no hCG in urine,
there is agglutination of latex particles (negative test).
This is commonly used as a slide test and requires only a
few minutes.
Sensitivity of agglutination inhibition test is >200
units/liter of hCG.
Radioimmunoassay, enzyme immunoassay, and
radioimmunometric assay are more sensitive and reliable
than agglutination inhibition assay.
Quantitative tests are employed for detection of very
early pregnancy, estimation of gestational age, diagnosis
of ectopic pregnancy, evaluation of threatened abortion,
and management of GTD.

REFERENCE RANGES
• Serum human chorionic gonadotropin:
– Non-pregnant females: <5.0 mIU/ml
– Pregnancy: 4 weeks after LMP: 5-100 mIU/ml
– 5 weeks after LMP: 200-3000 mIU/ml
– 6 weeks after LMP: 10,000-80,000 mIU/ml
– 7-14 weeks: 90,000-500,000 mIU/ml
Fig. 13.2: Principle of agglutination inhibition test for
– 15-26 weeks: 5000-80000 mIU/ml
diagnosis of pregnancy
– 27-40 weks: 3000-15000 mIU/ml
and are known as ‘heterophil’ antibodies. Fetal death,
abortion, dilute urine, and low sensitivity of a particular BIBLIOGRAPHY
test are causes of false-negative test. Renal failure leads 1. Burtis CA, Ashwood ER. Tietz fundamentals of clinical
to accumulation of interfering substances causing chemistry (5th Ed). Philadelphia: WB Saunders
incorrect results. Company, 2001.
In latex particle agglutination inhibition test 2. Davies S, Byrn F, Cole LA. Human chorionic gonado-
tropin testing for early pregnancy viability and
(Fig. 13.2), anti-hCG antibodies are incubated with complications. Clin Lab Med 2003;23:257-64.
patient’s urine. This is followed by addition of hCG- 3. Tenore JL. Ectopic pregnancy. Am Fam Physician
coated latex particles. If hCG is present in urine, anti- 2000;61:1080-8.
14

Infertility

Infertility refers to failure of a couple to achieve concep- MALE INFERTILITY


tion after one year of unprotected regular sexual
The male reproductive system consists of testes (paired
intercourse.
organs located in the scrotal sac that produce sperma-
In primary infertility, there is a failure of the couple
tozoa and secrete testosterone), a paired system of ducts
to conceive. In secondary infertility, there is a failure to
comprising of epididymis, vasa deferentia, and ejacu-
achieve conception after a previous pregnancy.
latory ducts (collect, store, and conduct spermatozoa),
About 80% of couples achieve conception within their
paired seminal vesicles and a single prostate gland
first year of attempt, another 10% will achieve within 2
years, while remaining 10% fail to conceive after 2 years. (produce nutritive and lubricating seminal fluid),
With time and therapeutic intervention, many subfertile bulbourethral glands of Cowper (secrete lubricating
couples can achieve a pregnancy and deliver a child. mucus), and penis (organ of copulation).
About 40% couples have primary, and 60% of couples The hypothalamus secretes gonadotropin releasing
have secondary infertility. hormone (GnRH) that regulates the secretion of the two
Source of infertility can be solely in female (30%), gonadotropins from the anterior pituitary: luteinizing
solely in male (30%), or in both partners (30%); the cause hormone (LH) and follicle stimulating hormone (FSH)
remains unknown in the remaining 10% cases even after (Fig. 14.1). Luteinizing hormone primarily stimulates the
extensive investigations. production and secretion of testosterone from Leydig

Fig. 14.1: Hypothalamus-pituitary-testis axis. + indicates stimulation; – indicates negative feedback


Infertility 151

cells located in the interstitial tissue of the testes.


Testosterone stimulates spermatogenesis, and plays a
role in the development of secondary sexual characters.
Testosterone needs to be converted to an important
steroidal metabolite, dihydrotestosterone within cells to
perform most of its androgenic functions. Testosterone
inhibits LH secretion by negative feedback. Follicle
stimulating hormone acts on Sertoli cells of seminiferous
tubules to regulate the normal maturation of the sperms.
Sertoli cells produce inhibin that controls FSH secretion
by negative feedback.
During sexual intercourse, semen is deposited into
the vagina. Liquefaction of semen occurs within 20-30
minutes due to proteolytic enzymes of prostatic fluid.
For fertilization to occur in vivo, the sperm must undergo
capacitation and acrosome reaction. Capacitation refers
to physiologic changes in sperms that occur during their
passage through the cervix of the female genital tract.
With capacitation, the sperm acquires (i) ability to
undergo acrosome reaction, (ii) ability to bind to zona
pellucida, and (iii) hypermotility. Sperm then travels
through the cervix and uterus up to the fallopian tube.
Binding of sperm to zona pellucida induces acrosomal
reaction (breakdown of outer plasma membrane by
enzymes of acrosome and its fusion with outer acrosomal
membrane, i.e. loss of acrosome). This is necessary for Fig. 14.2: Steps before and after fertilization of ovum
fusion of sperm and oocyte membranes. Acrosomal
reaction and binding of sperm and ovum surface proteins egg in the ampullary portion of the fallopian tube
is followed by penetration of zona pellucida of ovum by (Fig. 14.2).
the sperm. Following penetration by sperm, hardening
of zona pellucida occurs that inhibits penetration by Causes of Male Infertility
additional sperms. A sperm penetrates and fertilizes the Causes of male infertility are listed in Table 14.1.
Table 14.1: Causes of male infertility
1. Idiopathic
2. Hypothalamic-pituitary dysfunction (hypogonadotropic hypogonadism)
3. Testicular dysfunction:
• Radiation, cytotoxic drugs, antihypertensives, antidepressants
• General factors like stress, emotional factors, drugs like marijuana, anabolic steroids, and cocaine, alcoholism, heavy
smoking, undernutrition
• Mumps orchitis after puberty
• Varicocele (dilatation of pampiniform plexus of scrotal veins)
• Undescended testes (cryptorchidism)
• Endocrine disorders like diabetes mellitus, thyroid dysfunction
• Genetic disorders: Klinefelter’s syndrome, microdeletions in Y chromosome, autosomal Robertsonian translocation,
immotile cilia syndrome (Kartagener’s syndrome), cystic fibrosis, androgen receptor gene defect
4. Dysfunction of passages and accessory sex glands:
• Infections of epididymis: tuberculosis, gonorrhea, Chlamydia
• Congenital bilateral absence of vasa deferentia (cystic fibrosis), vasectomy
• Prostatitis
5. Dysfunction of sexual act:
• Impotence, erectile dysfunction
• Defects in ejaculation: retrograde (semen is pumped backwards in to the bladder), premature, or absent
• Hypospadias
152 Essentials of Clinical Pathology

Investigations of Male Infertility 4. Chromosomal analysis: This can reveal Klinefelter’s


syndrome (e.g. XXY karyotype) (Fig. 14.5), deletion
1. History: This includes type of lifestyle (heavy
in Y chromosome, and autosomal Robertsonian
smoking, alcoholism), sexual practice, erectile
translocation. It is necessary to screen for cystic
dysfunction, ejaculation, sexually transmitted
fibrosis carrier state if bilateral congenital absence of
diseases, surgery in genital area, drugs, and any vas deferens is present.
systemic illness. 5. Hormonal studies: This includes measurement of
2. Physical examination: Examination of reproductive FSH, LH, and testosterone to detect hormonal
system should includes testicular size, undescended abnormalities causing testicular failure (Table 14.2).
testes, hypospadias, scrotal abnormalities (like 6. Testicular biopsy: Testicular biopsy is indicated
varicocele), body hair, and facial hair. Varicocele can when differentiation between obstructive and non-
occur bilaterally and is the most common surgically
removable abnormality causing male infertility.
3. Semen analysis: See Chapter 15: Semen Analysis.
Evaluation of azoospermia is shown in Figure 14.3.
Evaluation of low semen volume is shown in Figure 14.4.

If all the values of semen analysis are normal


(according to WHO criteria), further testing is not
required. Further andrological tests are indicated only
if at least two semen examinations (done 1 month apart)
are abnormal. In patients with abnormal semen
analysis, the further basic laboratory tests done are
estimation of serum luteinizing hormone, follicle
stimulating hormone and testosterone.
Fig. 14.4: Evaluation of low semen volume

Fig. 14.3: Evaluation of azoospermia. FSH: Follicle stimulating hormone;


LH: Luteinizing hormone
Infertility 153

Fig. 14.5: Karyotype in Klinefelter’s syndrome (47, XXY)

Table 14.2: Interpretation of hormonal studies in male infertility

Follicle Luteinizing Testosterone Interpretation


stimulating hormone
hormone

Low Low Low Hypogonadotropic hypogonadism


(Hypothalamic or pituitary disorder)

High High Low Hypergonadotropic hypogonadism


(Testicular disorder)

Normal Normal Normal Obstruction of passages,


dysfunction of accessory glands
154 Essentials of Clinical Pathology

Box 14.1: Common initial investigations for


diagnosis of infertility

Males
• Semen analysis
• Blood glucose
• Endocrine tests: Serum FSH, LH, testosterone

Females
• Hemogram, X-ray chest, Mantoux test, erythrocyte
sedimentation rate
• Thyroid hormones, serum prolactin
• Endometrial biopsy
• Laparoscopy with hysteroscopy and/or hysterosalpin-
gography
• Transvaginal ultrsonography
Fig. 14.6: The hypothalamus-pituitary-ovarian axis

obstructive azoospermia is not evident (i.e. normal ovary collapses and fills with blood clot (corpus luteum).
FSH and normal testicular volume). LH converts granulose cells in the follicle to lutein cells
Common initial investigations for diagnosis of cause which begin to secrete progesterone. Progesterone
of infertility are shown in Box 14.1. stimulates secretion from the endometrial glands
(secretory phase) that were earlier under the influence
FEMALE INFERTILITY
of estrogen. Rising progesterone levels inhibit LH
The ovaries are the sites of production of female gametes production from the anterior pituitary. Without LH, the
or ova by the process of oogenesis. The ova are released corpus luteum regresses and becomes functionless
by the process of ovulation in a cyclical manner at regular corpus albicans. After regression of corpus luteum,
intervals. Ovary contains numerous follicles that contain production of estrogen and progesterone stops and
ova in various stages of development. During each
endometrium collapses, causing onset of menstruation.
menstrual cycle, up to 20 primordial follicles are activated
If the ovum is fertilized and implanted in the uterine
for maturation; however, only one follicle becomes fully
wall, human chorionic gonadotropin (hCG) is secreted
mature; this dominant follicle ruptures to release the
by the developing placenta into the maternal circulation.
secondary oocyte from the ovary. Maturation of the
follicle is stimulated by follicle stimulating hormone Human chorionic gonadotropin maintains the corpus
(FSH) secreted by anterior pituitary (Fig. 14.6). Maturing luteum for secetion of estrogen and progesterone till 12th
follicle secretes estrogen that causes proliferation of week of pregnancy. After 12th week, corpus luteum
endometrium of the uterus (proliferative phase). regresses to corpus albicans and the function of synthesis
Follicular cells also secrete inhibin which regulates of estrogen and progesterone is taken over by placenta
release of FSH by the anterior pituitary. Fall in FSH level till parturition.
is followed by secretion of luteinizing hormone (LH) by The average duration of the normal menstrual cycle
the anterior pituitary (LH surge). This causes follicle to is 28 days. Ovulation occurs around 14th day of the cycle.
rupture and the ovum is expelled into the peritoneal The time interval between ovulation and menstruation
cavity near the fimbrial end of the fallopian tube. The is called as luteal phase and is fairly constant (14 days)
fallopian tubes conduct ova from the ovaries to the (Fig. 14.7).
uterus. Fertilization of ovum by the sperm occurs in the
fallopian tube. Causes of Female Infertility
The ovum consists of the secondary oocyte, zona
pellucida and corona radiata. The ruptured follicle in the Causes of female infertility are shown in Table 14.3.
Infertility 155

Fig. 14.7: Normal menstrual cycle

Table 14.3: Causes of female infertility

1. Hypothalamic-pituitary dysfunction: 3. Dysfunction in passages


• Hypothalamic causes • Fallopian tubes
– Excessive exercise – Infections: Tuberculosis, gonorrhea, Chlamydia
– Excess stress – Previous surgery (e.g. laparotomy)
– Low weight – Tubectomy
– Kallman’s syndrome – Congenital hypoplasia, non-canalization
– Idiopathic – Endometriosis
• Pituitary causes • Uterus
– Hyperprolactinemia – Uterine malformations
– Hypopituitarism (Sheehan’s syndrome, – Asherman’s syndrome
Simmond’s disease) – Tuberculous endometritis
– Craniopharyngioma – Fibroid
– Cerebral irradiation • Cervix: Sperm antibodies
2. Ovarian dysfunction • Vagina: Septum
• Polycystic ovarian disease 4. Dysfunction of sexual act: Dyspareunia
(Stein-Leventhal syndrome)
• Luteinized unruptured follicle
• Turner’s syndrome
• Radiation or chemotherapy
• Surgical removal of ovaries
• Idiopathic

Investigations Anovulatory cycles are clinically characterized by


Evaluation of female infertility is shown in Figure 14.8. amenorrhea, oligomenorrhea, or irregular menstrua-
tion. However, apparently regular cycles may be
associated with anovulation.
Tests for Ovulation
2. Endometrial biopsy: Endometrial biopsy is done during
Most common cause of female infertility is anovulation. premenstrual period (21st-23rd day of the cycle). The
1. Regular cycles, mastalgia, and laparoscopic direct secretory endometrium during the later half of the
visualization of corpus luteum indicate ovulatory cycles. cycle is an evidence of ovulation.
156 Essentials of Clinical Pathology

Fig. 14.8: Evaluation of female infertility. FSH: Follicle stimulating hormone; LH: Luteinizing hormone;
DHEA-S: Dihydroepiandrosterone; TSH: Thyroid stimulating hormone; ↑ : Increased; ↓ : Decreased

3. Ultrasonography (USG): Serial ultrasonography is done


from 10th day of the cycle and the size of the
dominant follicle is measured. Size >18 mm is
indicative of imminent ovulation. Collapse of the
follicle with presence of few ml of fluid in the pouch
of Douglas is suggestive of ovulation. USG also is
helpful for treatment (i.e. timing of coitus or of
intrauterine insemination) and diagnosis of luteinized
unruptured follicle (absence of collapse of dominant
follicle). Transvaginal USG is more sensitive than
abdominal USG.
4. Basal body temperature (BBT): Patient takes her oral
temperature at the same time every morning before
arising. BBT falls by about 0.5 ° F at the time of
ovulation. During the second (progestational) half of Fig. 14.9: Ferning of cervical mucosa
the cycle, temperature is slightly raised above the pre-
ovulatory level (rise of 0.5° to 1°F). This is due to the
• Spinnbarkeit test: Cervical mucus is elastic and
slight pyrogenic action of progesterone and is
withstands stretching upto a distance of over 10
therefore presumptive evidence of functional corpus
cm. This phenomenon is called Spinnbarkeit or
luteum. the thread test for the estrogen activity. During
5. Cervical mucus study: the secretory phase, viscosity of the cervical mucus
• Fern test: During estrogenic phase, a characteristic increases and it gets fractured when stretched.
pattern of fern formation is seen when cervical This change in cervical mucus is evidence of
mucus is spread on a glass slide (Fig. 14.9). This ovulation.
ferning disappears after the 21st day of the cycle. 6. Vaginal cytology: Karyopyknotic index (KI) is high
If previously observed, its disappearance is during estrogenic phase, while it becomes low in
presumptive evidence of corpus luteum activity. secretory phase. This refers to percentage of super-
Infertility 157

ficial squamous cells with pyknotic nuclei to all Increased testosterone: Polycystic ovarian disease
mature squamous cells in a lateral vaginal wall smear. (PCOD), congenital adrenal hyperplasia (To differen-
Usually minimum of 300 cells are evaluated. The peak tiate PCOD from congenital adrenal hyperplasia,
KI usually corresponds with time of ovulation and ultrasound and estimation of dihydroepiandros-
terone or DHEA are done).
may reach upto 50 to 85.
3. Transvaginal ultrasonography: This is done for detection
7. Estimation of progesterone in mid-luteal phase (day 21 or
of PCOD.
7 days before expected menstruation): Progesterone
level > 10 nmol/L is a reliable evidence of ovulation Investigations to Assess Tubal and Uterine Status
if cycles are regular (Fig. 14.10). A mistimed sample 1. Infectious disease: These tests include endometrial
is a common cause of abnormal result. biopsy for tuberculosis and test for chlamydial IgG
antibodies for tubal factor in infertility.
Tests to Determine the Cause of Anovulation
2. Hysterosalpingography (HSG): HSG is a radiological
1. Measurement of LH, FSH, and estradiol during days 2 to contrast study for investigation of the shape of the
6: All values are low in hypogonadotropic hypo- uterine cavity and for blockage of fallopian tubes
gonadism (hypothalamic or pituitary failure). (Fig. 14.11). A catheter is introduced into the cervical
2. Measurement of TSH, prolactin, and testosterone if cycles canal and a radiocontrast dye is injected into the
are irregular or absent: uterine cavity. A real time X-ray imaging is carried
Increased TSH: Hypothyroidism
out to observe the flow of the dye into the uterine
Increased prolactin: Pituitary adenoma cavity, tubes, and spillage into the uterine cavity.
3. Hysterosalpingo-contrast sonography: A catheter is
introduced into the cervical canal and an echocontrast
fluid is introduced into the uterine cavity. Shape of
the uterine cavity, filling of fallopian tubes, and
spillage of contrast fluid are noted. In addition,
ultrasound scan of the pelvis provides information
about any fibroids or polycystic ovarian disease.
4. Laparoscopy and dye hydrotubation test with hysteroscopy:
In this test, a cannula is inserted into the cervix and
methylene blue dye is introduced into the uterine
cavity. If tubes are patent, spillage of the dye is
observed from the ends of both tubes. This technique
also allows visualization of pelvic organs, endo-
metriosis, and pelvic adhesions. If required, endo-
Fig. 14.10: Serum progesterone during metriosis and tubal blockage can be treated during
normal menstrual cycle the procedure.

Fig. 14.11: Hysterosalpingography


158 Essentials of Clinical Pathology

Possible pregnancy and active pelvic or vaginal • Serum dehydroepiandrosterone sulfate (DHEA-S):
infection are contraindications to tubal patency tests. – Adult males: 59-452 μg/ml
– Adult females: 12-379 μg/ml
REFERENCE RANGES BIBLIOGRAPHY
• Serum follicle stimulating hormone: 1. Brugh III, VM, Lipshultz LI. Male factor infertility: Evaluation
and management. Med Clin N Am 2004;88:367-85.
– Adult males: 1.4-15.4 mIU/ml 2. Burtis CA, Ashwood ER. Tietz fundamentals of clinical
– Adult females: Follicular phase: 1-10 mIU/ml; chemistry. 5th Ed. Philadelphia: WB Saunders
Ovulatory peak: 6-17 mIU/ml; Luteal phase: 1-9 Company, 2001.
mIU/ml 3. Dohle GR, Jungwirth A, Colpi G, Papp G, Pomerol J,
Hargreave TB. Guidelines on male infertility. European
• Serum luteinizing hormone: Association of Urology, 2006.
– Adult males: 1.24-7.8 mU/ml 4. Hirsh A. Male subfertility. BMJ 2003;327:669-72.
– Adult females: Follicular phase: 1.68-15 mIU/ml; 5. Hamilton-Fairley D, Taylor A. Anovulation. BMJ
Ovulatory peak: 21.9-56.6 mIU/ml; Luteal phase: 2003;327:546-9.
6. Jarrow JP. Diagnostic approach to the infertile male
0.61-16.3 mIU/ml
patient. Endocrinol Metab Clin N Am 2007;36:297-311.
• Serum testosterone: 7. Khalaf Y. Tubal subfertility. BMJ 2003;327:610-3.
– Adult males: 280-1100 ng/dl 8. Kolettis PN. Evaluation of the subfertile man. Am Fam
– Adult females: 15-70 ng/dl Physician 2003;67:2165-73.
9. Williams C, Giannopoulos T, Sherriff EA. Investigation
• Serum prolactin:
of infertility with the emphasis on laboratory testing
– Adult males: 3.0-14.7 ng/ml and with reference to radiological imaging. J Clin Pathol
– Adult females: 3.8-23.2 ng/ml 2003;56:261-7.
15

Semen Analysis

Semen (or seminal fluid) is a fluid that is emitted from Normal values for semen analysis are shown in Tables
the male genital tract and contains sperms that are 15.1 and 15.2.
capable of fertilizing female ova. Structures involved in
production of semen are (Box 15.1): INDICATIONS FOR SEMEN ANALYSIS
• Testes: Male gametes or spermatozoa (sperms) are
Availability of semen for examination allows direct
produced by testes; constitute 2-5% of semen volume.
examination of male germ cells that is not possible with
• Epididymis: After emerging from the testes, sperms
female germ cells. Semen analysis requires skill and
are stored in the epididymis where they mature;
should preferably be done in a specialized andrology
potassium, sodium, and glycerylphosphorylcholine
(an energy source for sperms) are secreted by laboratory.
epididymis. 1. Investigation of infertility: Semen analysis is the first
• Vas deferens: Sperms travel through the vas deferens step in the investigation of infertility. About 30% cases
to the ampulla which is another storage area. Ampulla of infertility are due to problem with males.
secretes ergothioneine (a yellowish fluid that reduces 2. To check the effectiveness of vasectomy by confir-
chemicals) and fructose (source of nutrition for ming absence of sperm.
sperms). 3. To support or disprove a denial of paternity on the
• Seminal vesicles: During ejaculation, nutritive and grounds of sterility.
lubricating fluids secreted by seminal vesicles and 4. To examine vaginal secretions or clothing stains for
prostate are added. Fluid secreted by seminal vesicles the presence of semen in medicolegal cases.
consists of fructose (energy source for sperms), amino 5. For selection of donors for artificial insemination.
acids, citric acid, phosphorous, potassium, and 6. For selection of assisted reproductive technology, e.g.
prostaglandins. Seminal vesicles contribute 50% to in vitro fertilization, gamete intrafallopian transfer
semen volume. technique.
• Prostate: Prostatic secretions comprise about 40% of
semen volume and consist of citric acid, acid COLLECTION OF SEMEN FOR
phosphatase, calcium, sodium, zinc, potassium, INVESTIGATION OF INFERTILITY
proteolytic enzymes, and fibrolysin.
Semen specimen is collected after about 3 days of sexual
• Bulbourethral glands of Cowper secrete mucus.
abstinence. Longer period of abstinence reduces motility
of sperms. If the period of abstinence is shorter than 3
Box 15.1: Contributions to semen volume days, sperm count is lower. The sample is obtained by
masturbation, collected in a clean, dry, sterile, and leak-
• Testes and epididymis: 10%
proof wide-mouthed plastic container, and brought to
• Seminal vesicles: 50%
the laboratory within 1 hour of collection. The entire
• Prostate: 40%
• Cowper’s glands: Small volume
ejaculate is collected, as the first portion is the most
concentrated and contains the highest number of sperms.
160 Essentials of Clinical Pathology

Table 15.1: Normal values of semen analysis (World Health Organization, 1999)

Test Result
1. Volume ≥2 ml
2. pH 7.2 to 8.0
3. Sperm concentration ≥20 million/ml
4. Total sperm count per ejaculate ≥40 million
5. Morphology ≥30% sperms with normal morphology
6. Vitality ≥75% live
7. White blood cells <1 million/ml
8. Motility within 1 hour of ejaculation
• Class A ≥25% rapidly progressive
• Class A and B ≥50% progressive
9. Mixed antiglobuiln reaction (MAR) test <50% motile sperms with adherent particles
10. Immunobead test <50% motile sperms with adherent particles

Table 15.2: Biochemical variables of semen analysis (World Helath Organization, 1992)

1. Total fructose (seminal vesicle marker) ≥13 μmol/ejaculate


2. Total zinc (Prostate marker) ≥2.4 μmol/ejaculate
3. Total acid phosphatase (Prostate marker) ≥200U/ejaculate
4. Total citric acid (Prostate marker) ≥52 μmol/ejaculate
5. α-glucosidase (Epididymis marker) ≥20 mU/ejaculate
6. Carnitine (Epididymis marker) 0.8-2.9 μmol/ejaculate

During transport to the laboratory, the specimen should


Box 15.2: Tests done on seminal fluid
be kept as close to body temperature as possible (i.e. by
carrying it in an inside pocket). Ideally, the specimen • Physical examination: Time to liquefaction, viscosity,
should be obtained near the testing site in an adjoining volume, pH, color
room. Condom collection is not recommended as it • Microscopic examination: Sperm count, vitality, motility,
morphology, and proportion of white cells
contains spermicidal agent. Ejaculation after coitus
• Immunologic analysis: Antisperm antibodies (SpermMAR
interruptus leads to the loss of the first portion of the test, Immunobead test)
ejaculate that is most concentrated; therefore this method • Bacteriologic analysis: Detection of infection
should not be used for collection. Two semen specimens • Biochemical analysis: Fructose, zinc, acid phosphatase,
should be examined that are collected 2-3 weeks apart; carnitine.
if results are significantly different additional samples • Sperm function tests: Postcoital test, cervical mucus
are required. penetration test, Hamster egg penetration assay, hypo-
osmotic swelling of flagella, and computer-assisted semen
analysis
EXAMINATION OF SEMINAL FLUID

The tests that can be done on seminal fluid are shown in Box 15.3: Semen analysis for initial
Box 15.2. Tests commonly done in infertility are shown investigation of infertility
in Box 15.3. The usual analysis consists of measurement • Volume
of semen volume, sperm count, sperm motility, and • pH
• Microscopic examination for (i) percentage of motile
sperm morphology.
spermatozoa, (ii) sperm count, and (iii) sperm morphology
Terminology in semen analysis is shown in Box 15.4.
Semen Analysis 161

(azoospermia) suggests obstruction of ejaculatory


Box 15.4: Terminology in semen analysis
ducts or absence of vas deferens. Low pH is usually
• Normozoospermia: All semen parameters normal associated with low semen volume (as most of the
• Oligozoospermia: Sperm concentration <20 million/ml volume is supplied by seminal vesicles).
(mild to moderate: 5-20 million/ml; severe: <5 million/ml)
• Azoospermia: Absence of sperms in seminal fluid Microscopic Examination
• Aspermia: Absence of ejaculate The most important test in semen analysis for infertility
• Asthenozoospermia: Reduced sperm motility; <50% of
is microscopic examination of the semen.
sperms showing class (a) and class (b) type of motility
OR <25% sperms showing class (a) type of motility.
Sperm Motility
• Teratozoospermia: Spermatozoa with reduced proportion
of normal morphology (or increased proportion of abnormal The first laboratory assessment of sperm function in a
forms) wet preparation is sperm motility (ability of the sperms
• Leukocytospermia: >1 million white blood cells/ml of to move). Sperm motility is essential for penetration of
semen
cervical mucus, traveling through the fallopian tube, and
• Oligoasthenoteratozoospermia: All sperm variables are
abnormal
penetrating the ovum. Only those sperms having rapidly
• Necrozoospermia: All sperms are non-motile or non-viable progressive motility are capable of penetrating ovum and
fertilizing it.
Principle: All motile and non-motile sperms are counted
Physical Examination in randomly chosen fields in a wet preparation under
40× objective. Result is expressed as a percentage of
Examination is carried out after liquefaction of semen
motile spermatozoa observed.
that occurs usually within 20-30 minutes of ejaculation.
1. Visual appearance: Normal semen is viscous and Method: A drop of semen is placed on a glass slide,
opaque gray-white in appearance. After prolonged covered with a coverslip that is then ringed with
abstinence, it appears slightly yellow. petroleum jelly to prevent dehydration, and examined
2. Viscosity: Immediately following ejaculation, normal under 40× objective. Atleast 200 spermatozoa are counted
semen is thick and viscous. It becomes liquefied in several different microscopic fields. Result is expressed
within 30 minutes by the action of proteolytic as a percentage of (a) rapidly progressive spermatozoa
enzymes secreted by prostate. If liquefaction does not (moving fast forward in a straight line), (b) slowly
occur within 60 minutes, it is abnormal. The viscosity progressive spermatozoa (slow linear or non-linear, i.e.
of the sample is assessed by filling a pipette with crooked or curved movement), (c) non-progressive
semen and allowing it to flow back into the container. spermatozoa (movement of tails, but with no forward
Normal semen will fall drop by drop. If droplets form progress), and (d) immotile spermatozoa (no movement
‘threads’ more than 2 cm long, then viscosity is at all) (WHO critera). Sperms of grades (c) and (d) are
increased. Increased semen viscosity affects sperm considered to be poorly motile (asthenospermia).
motility and leads to poor invasion of cervical mucus; Normally, ≥ 25% of sperms show rapid progressive
it results from infection of seminal vesicles or prostate. motility, or ≥ 50% of sperms show rapid progressive and
3. Volume: Volume of ejaculated semen sample should slow progressive motility.
normally be > 2 ml. It is measured after the sample If the proportion of motile spermatozoa is < 50%, then
has liquefied. Volume < 2.0 ml is abnormal, and is proportion of viable sperms should be determined by
associated with low sperm count. examining an eosin preparation.
4. pH: A drop of liquefied semen is spread on pH paper
(of pH range 6.4-8.0) and pH is recorded after 30 Sperm Viability or Vitality
seconds. Normal pH is 7.2 to 8.0 after 1 hour of Principle: A cell with intact cell membrane (a vital or
ejaculation. The portion of semen contributed by viable cell) will not take up the eosin Y and will not be
seminal vesicles is basic, while portion from prostate stained, while a non-viable or dead cell will have
is acidic. Low pH (< 7.0) with absence of sperms damaged cell membrane, will take up the dye, and will
162 Essentials of Clinical Pathology

Pasteur pipette. The chamber is then placed in a


humid box for 10-15 minutes for spermatozoa to
settle.
3. The chamber is placed on the microscope stage. Using
the 20× or 40× objective and iris diaphragm lowered
sufficiently to give sufficient contrast, number of
spermatozoa is counted in 4 large corner squares.
Spermatozoa whose heads are touching left and
upper lines of the square should be considered as
‘belonging’ to that square.
4. Sperm count per ml is calculated as follows:

Sperms counted × correction factor


for dilution
Sperm count = × 1000
Number of × Volume of
Fig. 15.1: Eosin-nigrosin stain. Dead sperms are stained squares counted 1 square
pink-red, while live sperms are stained white
Sperms counted × 20
be stained pink-red (Fig. 15.1). Another stain (e.g. = 1000
4 × 0.1
nigrosin) may be used to stain the background material.
The test is performed if motility is abnormal. = Sperms counted × 50, 000

Method
5. Normal sperm count is ≥ 20 million/ml (i.e. ≥ 20 ×
1. Mix one drop of semen with 1 drop of eosin-nigrosin
10 6/ml). Sperm count < 20 million/ml may be
solution and incubate for 30 seconds.
associated with infertility in males.
2. A smear is made from a drop placed on a glass slide.
3. The smear is air-dried and examined under oil- Sperm Morphology
immersion objective. White sperms are classified as
A smear is prepared by spreading a drop of seminal fluid
live or viable, and red sperms are classified as dead
on a glass slide, stained, and percentages of normal and
or non-viable. At least 200 spermatozoa are examined.
abnormal forms of spermatozoa are counted. The
4. The result is expressed as a proportion of viable
staining techniques used are Papanicolaou, eosin-
sperms against non-viable as an integer percentage.
nigrosin, hematoxylin-eosin, and Rose Bengal-toluidine
Seventy-five percent or more of sperms are normally
blue stain. Atleast 200 spermatozoa should be counted
live or viable.
under oil immersion. Percentages of normal and
Sperm Count abnormal spermatozoa should be recorded.
Normal morphology: A spermatozoon consists of three
Principle: The sperm count is done after liquefaction in a
main components: head, neck, and tail. Tail is further
counting chamber following dilution and the total
subdivided into midpiece, main (principle) piece, and
number of spermatozoa is reported in millions/ml (106/
end piece (Fig. 15.2 and Box 15.5).
ml).
Head is pear-shaped. Most of the head is occupied
Method by the nucleus which has condensed chromatin and few
1. Semen is diluted 1:20 with sodium bicarbonate- areas of dispersed chromatin (called nuclear vacuoles).
formalin diluting fluid (Take 1 ml liquefied semen in The anterior 2/3rds of the nucleus is surrounded by
a graduated tube and fill with diluting fluid to 20 ml acrosomal cap. Acrosomal cap is a flattened membrane-
mark. Mix well). bound vesicle containing glycoproteins and enzymes.
2. A coverslip is placed over the improved Neubauer These enzymes are required for separation of cells of
counting chamber and the counting chamber is filled corona radiata and dissolution of zona pellucida of ovum
with the well-mixed diluted semen sample using a during fertilization.
Semen Analysis 163

Box 15.5: Normal sperm morphology

• Total length of sperm: About 60 μ


• Head:
– Length: 3-5 μ
– Width: 2-3 μ
– Thickness: 1.5 μ
• Neck: Length: 0.3 μ
• Middle piece:
– Length: 3-5 μ
– Width: 1.0 μ
• Principal piece:
– Length: 40-50 μ
– Width: 0.5 μ
• End piece: 4-6 μ

Abnormal morphology (Fig. 15.3): WHO morphological


classification of human spermatozoa (1999) is given
below:
1. Normal sperm
2. Defects in head:
• Large heads
• Small heads
• Tapered heads
Fig. 15.2: Morphology of spermatozoa • Pyriform heads
• Round heads
• Amorphous heads
Neck is a very short segment that connects the head
• Vacuolated heads (> 20% of the head area occu-
and the tail. Centriole in the neck gives rise to axoneme
pied by vacuoles)
of the flagellum. Axoneme consists of 20 microtubules
• Small acrosomes (occupying < 40% of head area)
(arranged as a central pair surrounded by 9 peripheral
• Double heads
doublets) and is surrounded by condensed fibrous rings.
3. Defects in neck:
Middle piece is the first part of the tail and consists
• Bent neck and tail forming an angle >90° to the
of central axoneme surrounded by coarse longitudinal
long axis of head
fibers. These are surrounded by elongated mitochondria
4. Defects in middle piece:
that provide energy for movement of tail.
• Asymmetric insertion of midpiece into head
Principle or main piece constitutes most of the tail
• Thick or irregular midpiece
and is composed of axoneme that is surrounded by 9 • Abnormally thin midpiece
coarse fibers. This central core is surrounded by many 5. Defects in tail:
circularly arranged fibrous ribs. • Bent tails
Endpiece is the short tapering part composed of only • Short tails
axoneme. • Coiled tails
Normally, > 30% of spermatozoa should show normal • Irregular tails
morphology (WHO, 1999). The defects in morphology • Multiple tails
that are associated with infertility in males include • Tails with irregular width
defective mid-piece (causes reduced motility), an 6. Pin heads: Not to be counted
incomplete or absent acrosome (causes inability to 7. Cytoplasmic droplets
penetrate the ovum), and giant head (defective DNA • > 1/3rd the size of the sperm head
condensation). 8. Precursor cells: Considered abnormal
164 Essentials of Clinical Pathology

Fig. 15.3: Abnormal morphological sperm forms: (1) Normal sperm, (2) Large head, (3) Small head, (4) Tapered head, (5)
Pyriform head, (6) Round head, (7) Amorphous head, (8) Vacuoles in head, (9) Round head without acrosome, (10) Double
head, (11) Pin head, (12) Round head without acrosome and thick midpiece, (13) Coiled tail, and (14) Double tail

Round Cells a. SpermMARTM test: This test can detect IgG and IgA
antibodies against sperm surface in semen sample.
Round cells on microscopic examination may be white
In direct SpermMARTM IgG test, a drop each of semen
blood cells or immature sperm cells. Special stain
(peroxidase or Papanicolaou) is required to differentiate (fresh and unwashed), IgG-coated latex particles, and
anti-human immunoglobulin are mixed together on
between them. White blood cells >1 million/ml indicate
a glass slide. At least 200 motile spermatozoa are
presence of infection. Presence of large number of
examined. If the spermatozoa have antibodies on
immature sperm cells indicates spermatogenesis
their surface, antihuman immunoglobulin will bind
dysfunction at the testicular level.
IgG-coated latex particles to IgG on the surface of the
spermatozoa; this will cause attachment of latex
Immunologic Analysis
particles to spermatozoa, and motile, swimming
Antisperm Antibodies sperms with attached particles will be seen. If the
spermatozoa do not have antibodies on their surface,
The role of antisperm antibodies in causation of male
they will be seen swimming without attached
infertility is controversial. The immunological tests done
particles; the latex particles will show clumping due
on seminal fluid include mixed antiglobulin reaction to binding of their IgG to antihuman immuno-
(MAR test) and immunobead test. globulin.
The antibodies against sperms immobilize or kill In direct SpermMARTM IgA test, a drop each of fresh
them, thus preventing their passage through the cervix unwashed semen and of IgA-coated latex particles,
to the ovum. The antibodies can be tested in the serum, are mixed on a glass slide. The latex particles will
seminal fluid, or cervical mucus. If the antibodies are bind to spermatozoa if spermatozoa are coated with
present bound to the head of the sperm, they will prevent IgA antibodies.
the penetration of the egg by the sperm. If antibodies are In indirect SpermMAR TM tests, fluid without
bound to the tail of the sperm, they will retard motility. spermatozoa (e.g. serum) is tested for the presence of
Semen Analysis 165

antisperm antibodies. First, antibodies are bound to Postcoital (Sims-Huhner) Test


donor spermatozoa which are then mixed with the
This is the examination of the cervical mucus after coitus
fluid to be analyzed. These antibodies are then
and assesses the ability of the sperm to penetrate the
detected as described above for direct tests.
cervical mucus. The quality of the cervical mucus varies
Atleast 200 motile spermatozoa should be counted.
during the menstrual cycle, becoming more abundant
If >50% of spermatozoa show attached latex particles,
immunological problem is likely. and fluid at the time of ovulation (due to effect of
estrogen); this facilitates penetration of the mucus by the
b. Immunobead test: Antibodies bound to the surface of spermatozoa. Progesterone in the secretory phase
the spermatozoa can be detected by antibodies
increases viscosity of the mucus. Therefore cervical
attached to immunobeads (plastic particles with
mucus testing is scheduled just before ovulation
attached anti-human immunoglobulin that may be
(determined by basal body temperature records or
either IgG, IgA,or IgM). Percentage of motile
follicular sizing by ultrasonography). Postcoital test is
spermatozoa with attached two or more immuno-
beads are counted amongst 200 motile spermatozoa. the traditional method to detect the cervical factor in
Finding of >50% spermatozoa with attached beads is infertility. Cervical mucus is aspirated with a syringe
abnormal. shortly before the expected time of ovulation and 2-12
hours after intercourse. Gross and microscopic examina-
Biochemical Analysis of Semen tions are carried out to assess the quality of cervical
Biochemical markers (Table 15.2) can be measured in mucus (elasticity and drying pattern) and to evaluate the
semen to test the secretions of accessory structures. These number and motility of sperms (Box 15.6). If ≥ 10 motile
include fructose (seminal vesicles), zinc, citric acid or acid sperms are observed the test is considered as normal.
phosphatase (prostate), and α-glucosidase or carnitine An abnormal test may result from: (a) poor quality of
(epididymis). cervical mucus due to wrong judgment of ovulation,
cervicitis or treatment with antioestrogens (e.g. Clomid),
Test for Fructose and (b) absence of motile sperms due to ineffective
Resorcinol method is used for detection of fructose. In technique of coitus, lack of ejaculation, poor semen
this test, 5 ml of resorcinol reagent (50 mg resorcinol quality, use of coital lubricants that damage the sperm,
dissolved in 33 ml concentrated hydrochloric acid; dilute or presence of antisperm antibodies. Antisperm
up to 100 ml with distilled water) is added to 0.5 ml of antibodies cause immotile sperms, or agglutination or
seminal fluid. The mixture is heated and brought to boil. clumping of sperms; they may be present in either
If fructose is present, a red-colored precipitate is formed partner. If cervical factor is present, intrauterine
within 30 seconds. insemination is the popular treatment. The value of the
Absence of fructose indicates obstruction proximal postcoital test is disputed in the medical literature.
to seminal vesicles (obstructed or absent vas deferens) This test can be carried out if semen analysis is
or a lack of seminal vesicles. In a case of azoospermia, if normal, and the female partner is ovulating and fallopian
fructose is absent, it is due to the obstruction of tubes are not blocked. It is also done if antisperm
ejaculatory ducts or absence of vas deferens, and if
present, azoospermia is due to failure of testes to produce
sperm. Box 15.6: Interpretation of postcoital test
• Normal: Sperms are normal in amount and moving forward
Sperm Function Tests or Functional Assays
in the mucus; mucus stretches atleast 2 inches (5 cm)
These tests are available only in specialized andrology and dries in a fern-like manner.
laboratories. The tests are not standardized thus making • Abnormal: Absence of sperms or large number of sperms
interpretation difficult. If used singly, a sperm function are dead or sperms are clumped; cervical mucus cannot
stretch 2 inches (5 cm) or does not dry in a fern-like
test may not be helpful in fertility assessment. They are
manner.
more predictive if used in combination.
166 Essentials of Clinical Pathology

antibodies are suspected and male partner refuses semen Tests


analysis. 1. Microscopic examination for sperms: Presence of motile
sperms in vaginal fluid indicates interval of
Cervical Mucus Penetration Test < 8 hours. Smears prepared from collected samples
In this test, greatest distance traveled by the sperm in are stained and examined for the presence of sperms.
seminal fluid placed and incubated in a capillary tube 2. Acid phosphatase: Acid phosphatase is determined on
containing bovine mucus is measured. Majority of fertile vaginal or clothing samples. Due to the high level of
acid phosphatase in semen, its presence indicates
men show score >30 mm, while most infertile men show
recent sexual intercourse. Level of ≥50 U/sample is
scores <20 mm.
considered as positive evidence of semen.
Hamster Egg Penetration Assay 3. Determination of blood group substances: When semen
is positively identified in vaginal fluid or other
Hamster oocytes are enzymatically treated to remove the sample, test can be carried out for the presence of
outer layers (that inhibit cross-species fertilization). They blood group substances in the same sample. The
are then incubated with sperms and observed for ‘secretor’ individuals (80% individuals are secretors)
penetration rate. It can be reported as (a) Number of eggs will secrete the blood group substances in body fluids,
penetrated (penetration rate <15% indicates low fertility), including semen.
or as (b) Number of sperm penetrations per egg (Normal 4. Florence test: This test detects the presence of choline
>5). This test detects sperm motility, binding to oocyte, found in high concentration in semen. To several
and penetration of oocyte. There is a high incidence of drops of sample, add equal volume of reagent (iodine
false-negative results. 2.54 g, potassium iodide 1.65 g, distilled water 30 ml);
in positive test rhombic or needle-like crystals of
Hypo-osmotic Swelling of Flagella
periodide of choline form. False-positive tests can
This test assesses the functional integrity of the plasma occur due to high choline content of some other body
membrane of the sperm by observing curling of flagella fluids.
in hypo-osmotic conditions.
EXAMINATION OF SEMEN TO CHECK THE
Computer-assisted Semen Analysis EFFECTIVENESS OF VASECTOMY
Computer software measures various characteristics of The aim of post-vasectomy semen analysis is to detect
the spermatozoa; however, its role in predicting fertility the presence or absence of spermatozoa. The routine
potential is not confirmed. follow-up consists of semen analysis starting 12 weeks
(or 15 ejaculations) after surgery. If two successive semen
EXAMINATION FOR THE PRESENCE OF samples are negative for sperms, the semen is considered
SEMEN IN MEDICOLEGAL CASES as free of sperm. A follow-up semen examination at 6
months is advocated by some to rule out spontaneous
This includes examination of material obtained from reconnection.
vagina, stains from clothing, skin, hair, or other body
parts for semen. This is carried out in cases of alleged BIBLIOGRAPHY
rape or sexual assault. 1. Hirsh A. Male subfertility. BMJ 2003;327:669-72.
2. Phadke AM. Clinical Atlas of Sperm Morphology. New
Collection of Sample Delhi: Jaypee Brothers Medical Publishers (P) Ltd. 2007.
3. World Health Organization. Laboratory Manual for the
• Vagina: Direct aspiration or saline lavage Examination of Human Semen and Sperm-Cervical
• Clothing: When scanned with ultraviolet light, semen Mucus Interactions, 3rd edition. Cambridge: Cambridge
produces green white fluorescence. A small piece (1 University Press, 1992.
m2) of clothing from stained portion is soaked in 1-2 4. World Health Organization. WHO laboratory manual
for the examination of human semen and sperm-
ml of physiologic saline for 1 hour. A similar piece of
cervical mucus interaction. 4th edition. Cambridge, UK:
clothing distant from the stain is also soaked in saline New York, NY: Published on behalf of the World Health
as a control. Organization [by] Cambridge University Press; 1999.
Laboratory Hematology
16

Hematopoiesis

The physiologic process of formation of blood cells is


called as hematopoiesis (Greek haima: blood; poiesis: to
make). During 3rd week of embryonic life, hematopoiesis
begins in the yolk sac, as aggregates of blood cells called
as blood islands. Around 3rd month of fetal life, some of
the hematopoietic stem cells migrate to the liver which
then becomes the main site of hematopoiesis. Some blood
cell formation also occurs in the spleen, lymph nodes,
and thymus. Bone marrow starts producing blood cells
around 4th month of fetal life, and becomes the sole site
of hematopoiesis after birth (Box 16.1 and Fig. 16.1). In
certain conditions, hematopoiesis can become re-
established in liver, spleen, and lymph nodes; this is
called as extramedullary hematopoiesis.
Bone marrow is the soft, gelatinous tissue supported
by a reticulin framework that fills the intertrabecular
spaces in the cavities of the bones. There are two types Fig. 16.1: Sites of hematopoiesis
of bone marrow: red marrow composed of hematopoietic
tissue (active marrow) and yellow marrow composed of term pleuripotent refers to the ability to produce many
fat cells (inactive marrow). Active sites of hematopoiesis cell types. Most of the PHSCs are located in the bone
are: pelvis, vertebra, skull, ribs, sternum, and proximal marrow, but they are able to enter and circulate freely in
ends of long bones. In children, 100% of the total marrow peripheral blood. Stem cells have the ability of self-
space is active, while in adults, 50% of total marrow space renewal (to maintain a constant pool of undifferentiated
is active. cells) and differentiation. Hematopoietic stem cells are
All the blood cells circulating in the peripheral blood rare in the bone marrow (1 stem cell per 10000 to 1000000
are derived from the primitive mesenchymal cells called bone marrow cells). Primitive hematopoietic cells express
as pleuripotent hematopoietic stem cells (PHSCs); the CD34 antigen. Stem cells do not have any distinguishing
morphological features, and resemble a lymphocyte.
Box 16.1: Stages of hematopoiesis Stem cells can differentiate along different pathways and
produce other tissue cells (stem cell plasticity). Stem cells,
• Mesoblastic stage: Begins at 3rd week of gestation; as part of their proliferation, become committed to a
occurs in yolk sac. lineage and are then called as progenitor cells. Progenitor
• Hepatic stage: Begins 4-8 weeks of gestation; occurs cells do not have capacity of self-renewal, and have
in liver, thymus, and spleen
restricted multilineage potential.
• Myeloid stage: Begins at 12 weeks of gestation;
Two multipotential cell lines originate from the
occurs in cancellous areas of bones
pleuripotent hematopoietic stem cell: myeloid and
170 Essentials of Clinical Pathology

lymphoid and all blood cells belong to either of these Blood cells in the bone marrow can be considered to
two lineages. Myeloid cells include erythrocytes or red be in three compartments or pools: stem cell pool
cells, platelets, neutrophils, eosinophils, basophils, and (comprises of pleuripotent and multipotent stem cells,
monocytes. Lymphoid cells are of two main types: B and and commited CFUs), proliferating pool (morpholo-
T lymphocytes. The myeloid stem cell is called as CFU- gically identifiable precursors that are capable of DNA
GEMM (Colony forming unit granulocyte-erythrocyte- synthesis and mitosis), and storage pool (maturing and
monocyte-megakaryocyte). CFU refers to the develop- mature cells that are stored for later release in peripheral
ment of colonies of cells when marrow cells are injected blood).
in a tissue culture medium. CFU-GEMM leads to the Hematopoiesis is regulated by two main factors: (i)
formation of BFU-E (Burst Forming Unit-Erythroid), hematopoietic growth factors (HGFs) or cytokines,
CFU-Meg (Colony Forming Unit-Megakaryocyte), CFU- secreted by various cell types (Table 16.1), and (ii) stromal
GM (Colony Forming Unit-Granulocyte-Monocyte/ cells in the microenvironment of the bone marrow. HGFs
Macrophage), CFU-Eo (Colony Forming Unit- are glycoproteins secreted by various cell types which
Eosinophil), and CFU-Baso (Colony Forming Unit- regulate proliferation and differentiation of progenitor
Basophil). CFU-GM further produces CFU-G (Colony cells and function of mature blood cells. Stromal cells
Forming Unit-Granulocyte) and CFU-M (Colony secrete a variety of HGFs in the bone marrow micro-
Forming Unit-Monocyte/Macrophage) (Fig. 16.2). The environment; secondly hematopoietic progenitor cells
progenitor cells up to these stages are not identifiable adhere to receptors on stromal cells and on the stromal
morphologically; they can however be identified by their matrix that facilitates interaction of progenitor cells with
surface receptors. regulatory cytokines.

Fig. 16.2: Hierarchy of hematopoiesis. Abbreviations: SCF: Stem cell factor; IL: Interleukin; CFU-GEMM: Colony forming unit
granulocyte-erythrocyte-monocyte-megakaryocyte; CFU-GM: Colony forming unit granulocyte-macrophage; TPO: Thrombopoietin;
CFU- G: Colony forming unit granulocyte; CFU-M: Colony forming unit macrophage; CFU-Eo: Colony-forming unit eosinophil;
CFU-Baso: Colony forming unit basophil; CFU-Meg: Colony forming unit megakaryocyte; Epo: Erythropoietin; GM-CSF: granulocyte
macrophage colony stimulating factor; G-CSF: Granulocyte colony stimulating factor; M-CSF: Macrophage Colony stimulating
factor
Hematopoiesis 171

Table 16.1: Hematopoietic growth factors

Growth factor Site of synthesis Site of action

1. Granulocyte macrophage T cells, fibroblasts, Granulocytes, monocytes,


colony stimulating factor endothelial cells platelets, red cells
2. Granulocyte colony Monocytes, fibroblasts Neutrophils
stimulating factor
3. Macrophage colony Monocytes, endothelial Monocytes
stimulating factor cells, fibroblasts
4. Erythropoietin Kidney Red cells
5. Thrombopoietin Liver Platelets
6. Stem cell factor — Pleuripotent stem cells
7. Interleukin 3 T cells Hematopoietic stem cells
8. Interleukin 5 T cells Eosinophils
9. Interleukin 6 T lymphocytes, B and T cells
monocytes, fibroblasts

Fig. 16.3: Stages of erythropoiesis

Table 16.2: Stages in maturation of red blood cells


Cell Size (μ) Nucleus Cytoplasm

1. Proerythroblast 15-20 Large with immature chromatin, Blue


usually a single nucleolus
2. Early erythroblast 12-16 Fine chromatin clumps, nucleolus Deep blue due to high RNA
(Basophilic) barely visible
3. Intermediate erythroblast 12-15 Smaller size, chromatin clumps Polychromatic due to
(Polychromatic) coarse beginning of
hemoglobinization
4. Late erythroblast 8-12 Small, dense, pyknotic, and Polychromatic
(Orthochromatic) eccentric
5. Reticulocyte (Polychromatic 8-10 No nucleus Polychromatic, remnants of
red cell) RNA visible as a network on
supravital staining
6. Erythrocyte 7-8 No nucleus Pink

RED BLOOD CELLS identifiable erythroid precursor. It proliferates to


generate sequentially basophilic, polychromatic, and
Erythropoiesis
orthochromatic erythroblast; these cells are named after
Stages of erythropoiesis are shown in Figure 16.3 and staining characteristics of cytoplasm. Reticulocytes still
Table 16.2. Proerythroblast is the earliest morphologically retain some cytoplasmic organelles (a fine reticulim of
172 Essentials of Clinical Pathology

ribosomal RNA). With each stage of development, cell Red Cell Enzymes
size and nuclear size become smaller, chromatin
Energy for various metabolic processes in the red cell
clumping increases, and ultimately nucleus is extruded.
(transport of oxygen and carbon dioxide, membrane
Color of cytoplasm gradually changes from basophilic
pump, prevention of oxidant damage to hemoglobin, and
to pink due to hemoglobinization. A mature red cell or
erythrocyte is a biconcave, non-nucleated disk of 7-8 μ to convert methemoglobin to oxyhemoglobin) is
size. provided by glycolysis or Embden-Meyerhof pathway
and hexose monophosphate (HMP) shunt. A metabolic
Hemoglobin shunt in glycolytic pathway is Rapoport-Luebering shunt
Hemoglobin (average molecular weight 64,000) consists for generation of 2,3-diphosphoglycerate (2,3-DPG), an
of heme (iron and protoporphyrin) and globin (two important determinant for oxygen affinity of hemo-
polypeptide chains). A heme group is attached to each globin. In HMP pathway, NADPH is produced that
polypeptide chain. Normally, different types of hemo- reduces oxidized glutathione; reduced glutathione along
globins are present during embryonic life, fetal life, with glutathione peroxidase detoxifies hydrogen
infancy, and adulthood (Table 16.3). peroxide and prevents oxidant damage to hemoglobin.
Hemoglobin (Hb) Gower I, Hb Gower II, and Hb The enzyme glucose 6 phosphate dehydrogenase (G6PD)
Portland predominate during embryonic life, while Hb is necessary for generation of NADPH (Fig. 16.5).
F predominates during fetal life. HbF starts to decline
after 36 weeks of gestation, and constitutes <1% of total Red Cell Membrane
hemoglobin in adults. HbA becomes the main hemo-
globin after 3-4 months of age. Red cell cytoskeletal proteins lie beneath the lipid bilayer
Steps in the synthesis of β globin chain of hemo- and consist of horizontally oriented spectrin hetero-
globin A are shown in Figure 16.4. dimers (α and β spectrin intertwined together) that link
Biosynthesis of heme is outlined in Chapter 31: with ankyrin, proteins 4.1 and 4.2, and band 3 (vertical
Laboratory Tests in Porphyrias. elements) (Fig. 16.6). Cytoskeletal proteins maintain
The function of red cells is transport of oxygen and shape and allow flexibility permitting red cells to traverse
carbon dioxide. Oxygen binds covalently and reversibly through the capillaries of small diameter.
with hemoglobin, with maximum 4 oxygen molecules The lifespan of red cells is about 120 days, after which
binding to four iron molecules (i.e. one molecule of they are ingested and degraded by mononuclear
oxygen binds to each heme group). Oxygen affinity of phagocytes. Iron and amino acids are recycled for reuse,
hemoglobin is affected by pH, temperature, and oxygen while the remainder of the heme molecule is metabolized
pressure in capillaries. to bilirubin and excreted by liver in bile.

Table 16.3: Types of hemoglobin during different periods of life

Type of hemoglobin Globin chains Period of life


when predominant

Hemoglobin Gower I ζ 2ε2 Embryonic


Hemoglobin Gower II α 2ε2 Embryonic
Hemoglobin Portland ζ 2γ2 Embryonic
Hemoglobin F (Fetal hemoglobin) α 2γ2 Fetal
Hemoglobin A (Adult hemoglobin) α 2β2 Adult
Hemoglobin A2 α 2δ2 Adult
Hematopoiesis 173

Fig. 16.4: (A) β globin gene cluster on chromosome 11; (B) Steps in the synthesis of β globin
polypeptide chain from β globin gene

Fig. 16.5: Role of glucose-6-phosphate dehydrogenase enzyme in detoxification of toxic peroxide. Abbreviations: HK: Hexokinase;
GSSG: Oxidized glutathione; GSH: Reduced glutathione; H2O2: Hydrogen peroxide; 6PGL: 6 Phosphogluconolaconase; 6PGD:
6 Phosphogluconate dehydrogenase; Ru5PI: Ribulose 5 phosphate isomerase
174 Essentials of Clinical Pathology

Fig. 16.6: Structure of red cell membrane

WHITE BLOOD CELLS Basophils


Neutrophils Stages in the maturation of basophils are similar to
Morphologically identifiable stages in maturation of neutrophils. Basophils are 9-12 μ in size, and their
neutrophils are shown in Table 16.4 and Figure 16.7. cytoplasm is filled with coarse deep purple-black
Neutrophils play an important role in acute infla- granules that obscure the nucleus. Nucleus is segmented
mmation. into 2-3 lobes. The basophil granules contain histamine
and heparin. Basophils play a role in allergic and
Eosinophils anaphylactic reactions.
Various stages in maturation of eosinophils are similar
Monocytes
to neutrophils with primary or specific granules first
appearing in myelocyte stage. The mature eosinophil (15- The maturation sequence is monoblast, promonocyte,
16 μ) is slightly larger than a neutrophil, usually bilobed, and monocyte (Fig. 16.8). Monocytes are the largest white
and contains numerous bright orange-red granules. The blood cells in peripheral blood (15-20 μ) with irregular
eosinophil granules contain major basic protein that is shape, oval or kidney-shaped nucleus, and fine reticular
toxic to many parasites. chromatin. Cytoplasm is abundant, blue-gray with

Table 16.4: Stages in maturation of neutrophils


Cell Size (μ) Nucleus Cytoplasm

1. Myeloblast 15-20 Large, immature, fine dispersed chromatin, 2-5 nucleoli Scanty, light blue
2. Promyelocyte 16-20 Similar to myeloblast, slightly eccentric Azurophil granules
3. Myelocyte 14-18 Chromatin condensed, no nucleoli Specific granules
predominate
4. Metamyelocyte 14-18 Indented or kidney-shaped; peripheral clumping Faint pink; numerous
of chromatin specific granules
5. Band form 14-16 U-shaped or band-like nucleus with heavily Pink; numerous
clumped chromatin specific granules
6. Neutrophil 14-15 2-5 lobes joined by chromatin strands Pink; numerous
specific granules
Hematopoiesis 175

sible for cell-mediated immunity; they also regulate


humoral response. The T lymphocyte precursor cell can
differentiate into (i) T helper inducer cells which
differentiate into TH1 and TH2 cells, (ii) T suppressor
cells, and (iii) cytotoxic T cells.
Natural killer cells comprise about 10-15% of
lymphocytes in peripheral blood. NK cells can attack and
kill virus-infected cells and some neoplastic cells without
previous sensitization.
Features of B and T lymphocytes are compared in
Table 16.5.
B cell development (B cell ontogeny): There are two stages
of B cell development: antigen-independent (occurring
Fig. 16.7: Stages of granulopoiesis in bone marrow) and antigen-dependent (occurring in
peripheral lymphoid organs). Heavy chain gene
rearrangement is followed by rearrangement of light
chain genes. Following expression of TdT and HLA-DR,
there is sequential appearance of antigens on the surface
of developing B cells: CD19, CD10, and CD20. Plasma
cells express CD38 antigen.
T cell development (T cell ontogeny): Pre-T cells are
transported from the bone marrow to the thymus, where
maturation occurs. Initially the immature thymocytes
express CD7, TdT, and cytoplasmic CD3. Subsequently,
Fig. 16.8: Formation of monocytes both CD4 and CD8 antigens are acquired; with matu-
ration, either CD4 or CD8 is retained.
Normal mature white blood cells in peripheral blood
ground glass appearance and often contains fine are shown in Figure 16.9.
azurophil granules and vacuoles. After migration to
tissues, they are called as macrophages. Monocytes Human Leukocyte Antigens
function as phagocytic cells, antigen presenting cells, and Human leukocyte antigens or HLA (so called because
also produce a variety of cytokines. they were first discovered on white blood cells) are any

Lymphocytes
There are three main types: B lymphocytes, T lympho-
cytes, and natural killer (NK) cells.
B lymphocytes comprise about 10-20% of lympho-
cytes in peripheral blood. In lymphoid organs they are
located in superficial cortex, germinal centers, and mantle
zone of lymph nodes, follicles in spleen, and mucosa-
associated lymphoid tissue (MALT) in gastrointestinal
and respiratory tracts. On antigen stimulation, B
lymphocytes differentiate into plasma cells that produce
immunoglobulins (antibodies).
T lymphocytes comprise 60-70% of circulating
lymphocytes. In lymphoid organs, they are located in
thymus, paracortex of lymph nodes, and periarteriolar Fig. 16.9: Normal mature white blood cells in
lymphoid sheaths in spleen. T lymphocytes are respon- peripheral blood
176 Essentials of Clinical Pathology

Table 16.5: Comparison of T and B lymphocytes

Feature T lymphocytes B lymphocytes

1. Origin Stem cells in bone marrow Stem cells in bone marrow


2. Maturation Thymus Bone marrow, lymphoid organs
3. Name derivation Thymus-derived, meaning Bone marrow-derived (however, the B
they mature in thymus stands for bursa of Fabricius, a lymphoid
organ of birds in which B cells were first
discovered)
4. Lifespan Long-lived (2-4 years) Short-lived (weeks to a few months)
5. Differentiation Helper, suppressor, and Plasma cells
cytotoxic cells
6. Function Cell-mediated immunity, Humoral immunity (Secretion of antibodies
regulation of function of B cells that circulate through humors or body fluids)
7. Surface receptor T cell receptor Immunoglobulin
8. Fc and C3 receptor Absent Present
9. Electron microscopy Smooth surface Microvilli
10. Peripheral blood (%) 60-70% 10-20%
11. Location in lymph Paracortex Germinal centers of follicles, medullary
nodes cords
12. Location in spleen Periarteriolar lymphoid sheaths Germinal centers of follicles
13. Immunological markers CD1, CD2, CD3, CD4/CD8, CD5, CD19, CD20, CD21, CD22
CD6, CD7

of the numerous antigens that are encoded by a cluster


of genes on short arm of chromosome 6 (6p21) called as
major histocompatibility complex, that are extremely
polymorphic (Fig. 16.10). Each gene has an unusually
large number of alleles, so that it is very rare for two
individuals to have the same set of HLA molecules.
Alleles at the MHC loci are inherited in a codominant
Mendelian manner. HLA antigens are of three main
types: classes I, II, and III.
Class I HLA antigens: These include HLA-A, HLA-B, and
HLA-C with numerous alleles. They are associated non-
covalently with β2-microglobulin. They are expressed on
the surface of all nucleated cells. Their major function is
presentation of antigen to cytotoxic T cells. Class I
antigens are detected by lymphocytotoxicity test.
Class II antigens: These include HLA-DR, HLA-DQ, and
HLA-DP with numerous alleles. These consist of two
glycoprotein chains (α and β) bound non-covalently.
They are expressed primarily on antigen-presenting cells
like macrophages, B lymphocytes, and activated T Fig. 16.10: MHC complex (above) and HLA
lymphocytes. Their major function is antigen presen- antigens (below)
Hematopoiesis 177

tation to T helper lymphocytes. Mixed lymphocyte lymphocyte-antiserum mixture. If corresponding


culture or mixed lymphocyte reaction is used for antigen is present on lymphocytes, cell damage and
detection of class II antigens. Primed lymphocyte typing death occurs due to antigen-antibody reaction
is used for detection of HLA-DP antigens. followed by complement activation. Dead and live
cells are differentiated by staining with a vital dye
Class III antigens: There are several class III genes that
(dead cells stain while live cells remain unstained). If
encode certain complement components (C2, C4, and
≥ 60% cells are stained, they are considered to be
Factor B) and cytokines (tumor necrosis factor).
positive for the particular HLA antigen.
2. Mixed lymphocyte culture or mixed lymphocyte reaction:
Importance of HLA antigens
Lymphocytes from two different individuals are
• Organ transplantation: The immune system relies on obtained. Lymphocytes from one individual are
HLA antigens to distinguish self from nonself. Kidney inactivated to suppress their division; these lympho-
and bone marrow transplantation depend heavily on cytes are called stimulator cells. The lymphocytes
HLA antigen typing since these tissues express HLA from two individuals are cultured together. If
antigens to a great extent. antigens on stimulator cells are foreign to the
• Recognition of foreign antigens and immunity responder lymphocytes (i.e. to lymphocytes from
• Association between certain HLA antigens and other individual), responder cells show proliferation
diseases like ankylosing spondylitis, type 1 diabetes and synthesis of DNA (blastogenic response). DNA
mellitus, systemic lupus erythematosus, myasthenia synthesis is quantitated by incorporation of 3thymi-
dine.If antigens on both responder and stimulator
gravis, and Sjogren’s syndrome.
cells are similar there is no blastogenic response.
• Paternity testing.
3. Primed lymphocyte typing: This is a modification of
Methods for HLA Antigen Typing mixed lymphocyte culture. The culture of lympho-
cytes is extended during which stimulator cells die
Serological Testing and responder cells cease to proliferate. When
1. Lympocytotoxicity test (Fig. 16.11): Lymphocytes are responder cells are re-exposed to the same antigen-
separated from blood by density gradient centri- bearing cells as stimulator cells, rapid proliferation
of responder cells occurs.
fugation. Lymphocytes are then added to known
specific antisera (containing known specific anti- Molecular Genetic Techniques
bodies to HLA antigens). Complement is added to
1. Restriction fragment length polymorphism: This method
relies on recognition of certain DNA nucleotide
sequences by restriction enzymes and cutting the
DNA at these points. For example, DNA for one HLA
antigen will have a different restriction site as
compared to DNA for another HLA antigen so that
fragments of different sizes are obtained with the
same enzyme which can be determined by electro-
phoresis.
2. Polymerase chain reaction (PCR): Various steps in this
technique are: (1) DNA is obtained from the nuclei of
white blood cells; (2) Double-stranded DNA is
denatured into single-stranded DNA; (3) An oligo-
nucleotide primer sequence (‘probe’) complementary
to a small segment of only one HLA allele is chosen,
that will attach to single stranded DNA; (3) After
attachment, that part of DNA becomes double
stranded (after addition of DNA polymerase and
deoxyrionucleotide triphosphates); (4) Thus two
Fig. 16.11: Principle of lymphocytotoxicity test copies of DNA of interest are obtained; the double
178 Essentials of Clinical Pathology

stranded DNA is again denatured and associated


with primers to yield four copies; the cycle is repeated
to obtain sufficient amount of DNA to be elctro-
phoresed and typed. (5) If any DNA is detected on
agarose gel electrophoresis, particular HLA is present.
A number of PCR-based methods are used like
sequence specific priming and sequence specific
oligonucleotide priming.

THROMBOPOIESIS
Platelets are produced by cytoplasmic fragmentation of Fig. 16.12: Stages of platelet formation. The morphologically
large cells in bone marrow called as megakaryocytes, recognizable stages are megakaryoblast (high nucleo-
cytoplasmic ratio with basophilic cytoplasm), promegakaryocyte
which in turn are derived from megakaryoblasts. The
(nucleus typically horse-shoe shaped), and megakaryocyte
morphologically identifiable stages in thrombopoiesis (cytoplasm granular, nucleus highly lobular). Megakaryocytes
are: megakaryoblast, promegakaryocyte, megakaryocyte, undergo endomitosis and are the largest cells in the bone
marrow
and platelets (Fig. 16.12). Although nucleus of a mega-
karyocyte divides, cell division does not occur so that a
very large cell with multiple nuclear lobes (increased cytoplasmic processes through the sinusoidal wall in
ploidy) is formed (the process whereby nuclear DNA is marrow and platelets are released directly in bloodstream
duplicated without cell division is called as endomitosis). by fragmentation of cytoplasm. The lifespan of platelets
Megakaryocyte is the largest normally occurring hemato- is about 7-10 days.
poietic cell in the bone marrow. Each megakaryocyte
BIBLIOGRAPHY
produces about 4000 platelets during its lifespan. The
1. Hoffbrand AV, Moss PAH, Pettit JE (Eds). Essential
primary regulator of megakaryocyte differentiation is Hematology (5th Ed). Blackwell Publishing Ltd, 2006.
thrombopoietin (secreted by liver and stromal cells of 2. Kawthalkar SM. Essentials of Hematology. New Delhi:
bone marrow). A mature megakaryocyte extends Jaypee Brothers Medical Publishers (P) Ltd, 2006.
17

Collection of Blood

For reliable and accurate results of laboratory tests, it is


essential to follow a standard procedure for specimen
collection. For hematological investigations, blood
sample can be obtained from the skin puncture or
venepuncture.

SKIN PUNCTURE
This method is commonly used in infants and small
children and if the amount of blood required is small. It
is suitable for cell counts, estimation of hemoglobin,
determination of hematocrit by micro method, and
preparation of blood films.
Blood obtained by skin puncture is also called as
capillary blood. However, it is a mixture of blood from
capillaries, venules, and arterioles. It also contains some
tissue fluid.
In adults, blood is obtained from the side of a ring or
middle finger (distal digit) or ear lobe. In infants, it is
collected from the heel (lateral or medial aspect of plantar Fig. 17.1: (A) Blood lancet and sites of (B) finger
surface) or great toe (Fig. 17.1). puncture (cross) and (C) heel puncture (shaded areas)
The puncture site is cleansed with 70% ethanol or
other suitable disinfectant. After drying, a puncture,
VENOUS BLOOD COLLECTION
sufficiently deep to allow free flow of blood, is made with
a sterile, dry, disposable lancet. The first drop of blood is When multiple tests are to be done and larger quantity
wiped away with sterile, dry cotton as it contains tissue of blood is needed, anticoagulated venous blood should
fluid. Next few drops of blood are collected. Excessive be obtained.
squeezing should be avoided, as it will dilute the blood
Method
with tissue fluid. After collection a piece of sterile cotton
is pressed over the puncture site till bleeding ceases. 1. Due to the ease of access, blood is best obtained from
As compared to the venous blood, hemoglobin, the veins of the antecubital fossa (Fig. 17.2). A rubber
hematocrit, and red cell count are slightly higher in blood tourniquet (18 inches long × 3/4 or 1 inch in adults
from skin puncture. As platelets adhere to the puncture and 12 inches × 1/8 inch in children) is applied to
site, platelet count is lower. Because of small sample size, the upper arm. It should not be too tight and should
immediate repeat testing is not possible if the result is not remain in place for more than two minutes.
abnormal. Patient is asked to make a fist so that veins become
Blood should not be collected from cold, cyanosed more prominent and palpable.
skin since false elevation of values of hemoglobin and 2. Venepuncture site is cleansed with 70% ethanol and
red/white cell counts will be obtained. allowed to dry.
180 Essentials of Clinical Pathology

Check whether the patient is feeling faint and


bleeding has stopped. Cover the puncture site with an
adhesive bandage strip.
After use, disposable needles should be placed in a
puncture-proof container for proper disposal. Recapping
of needle by hand can cause needle-stick injury.
The container is labeled. Time of collection should be
noted on the label. Sample should be sent immediately
to the laboratory with accompanying properly filled
order form.

Precautions
1. Blood is never collected from an intravenous line or
from the arm being used for intravenous line (since
it will dilute the blood sample). Blood is not collected
from a sclerosed vein and from an area with hema-
toma.
2. Tourniquet should not be too tight and should not be
Fig. 17.2: Common sites of venepuncture in antecubital applied for more than 2 minutes as it will cause
fossa (red circles) hemoconcentration and alteration of test results.
3. Puncture site should be allowed to dry completely
after cleaning with alcohol (before performing the
3. The selected vein is anchored by compressing and venepuncture).
pulling the soft tissues below the puncture site with 4. Tourniquet should be released before removing the
the left hand.
needle from the vein (to prevent hematoma
4. Sterile, disposable needles and syringes should be
formation).
used for venepuncture. Needle size should be 19- to
5. To avoid hemolysis, blood is withdrawn gradually, a
21-gauge in adults and 23-gauge in children.
Venepuncture is performed with the bevel of the small-bore needle should not be used, and the needle
needle up and along the direction of the vein. Blood is detached from the syringe before dispensing blood
is withdrawn slowly. Pulling the plunger quickly can into the container.
cause hemolysis and collapse of the vein. 6. All blood samples are considered as infectious and
Tourniquet should be released as soon as the blood proper precautions should be observed while
begins to flow into the syringe. collecting blood either from a vein or a skin puncture.
5. When the required amount of blood is withdrawn, Anticoagulated blood sample should be tested within
the patient is asked to open his/her fist. The needle 1-2 hours of collection. If this is not possible, sample can
is withdrawn from the vein. A sterile cotton gauze is be stored in a refrigerator at 4-6°C for maximum of 24
pressed over the puncture site. Patient is asked to hours. After the sample is taken out of refrigerator, it
press the gauze over the site till bleeding stops. should be allowed to return to room temperature, mixed
6. The needle is detached from the syringe and the properly, and then tested.
required amount of blood is carefully delivered into
the tube containing appropriate anticoagulant (see Complications
later). If the blood is forced through the needle 1. Failure to obtain blood: This happens if vein is missed,
without detaching it, hemolysis can occur. Containers or excessive pull is applied to the plunger causing
may be glass bottles or disposable plastic tubes with collapse of the vein.
caps and flat bottom. 2. Occurrence of hematoma, thrombosis, thrombo-
7. Blood is mixed with the anticoagulant in the container phlebitis, abscess, or bleeding.
thoroughly by gently inverting the container several 3. Transmission of infections like hepatitis B or human
times. The container should not be shaken vigorously immunodeficiency virus if reusable needles and
as it can cause frothing and hemolysis. syringes, which are not properly sterilised, are used.
Collection of Blood 181

ANTICOAGULANTS
Anticoagulants used for hematological investigations are
ethylene diamine tetra-acetic acid (EDTA), heparin,
double oxalate, and trisodium citrate (Table 17.1).

Ethylene Diamine Tetra-acetic Acid (EDTA)


This is also called as Sequestrene or Versene. This is the
recommended anticoagulant for routine hematological
investigations. However, it cannot be used for coagu-
lation studies. Disodium and dipotassium salts of EDTA
are in common use. International Committee for
Standardization in Hematology recommends dipo-
tassium EDTA since it is more soluble. It is used in a Fig. 17.3: Changes in blood cell morphology (crenation of red
concentration of 1.5 mg/ml of blood. Dried form of cells, separation of nuclear lobes of neutrophil, vacuoles in
anticoagulant is used as it avoids dilution of sample. Its cytoplasm, and irregular lobulation of monocyte and lymphocyte
mechanism of action is chelation of calcium. nuclei) due to storage of blood in EDTA anticoagulant for
Proportion of anticoagulant to blood should be prolonged time
maintained. EDTA in excess of 2 mg/ml causes shrinkage
of and degenerative changes in red and white blood cells,
decrease in hematocrit, and increase in mean corpuscular Box 17.1: Changes occurring due to prolonged storage
hemoglobin concentration. Excess EDTA also causes of blood in EDTA
swelling and fragmentation of platelets, which leads to • Blood smear:
erroneously high platelet counts. – Red cells: Crenation and spherocytic change
– Neutrophils: Separation of nuclear lobes, loss of
Prolonged storage of blood in EDTA anticoagulant
granules, vacuoles in cytoplasm
leads to alterations as shown in Figure 17.3 and Box 17.1. – Monocytes and lymphocytes: Irregular lobulation
EDTA is used for estimation of hemoglobin, hema- of nucleus, vacuoles in cytoplasm
tocrit, cell counts, making blood films, sickling test, – Platelets: Disintegration
reticulocyte count, and hemoglobin electrophoresis. • Hematological tests:
– Packed cell volume and mean cell volume:
Preparation Increase due to red cell swelling
– Osmotic fragility: Increases
Dipotassium ethylene 20 gm – Total leucocyte and platelet counts: Progressive
diamine tetra-acetic acid decrease
Distilled water 200 ml

Table 17.1: Salient features of three main anticoagulants used in the hematology laboratory

Name Mode of action Concentration Main uses Disadvantages

1. EDTA Chelation of calcium 1.5 mg/ml Complete blood Cannot be used for
count, blood smear, coagulation tests; can cause
hemoglobin pseudothrombocytopenia on
electrophoresis, hematology analyzer
sickling test
2. Heparin Inhibition of thrombin 15 U/ml Osmotic fragility test, Causes blue background of
activity immunophenotyping blood smears; expensive
3. Sodium citrate Chelation of calcium 0.109 mg/ml Coagulation studies, Liquid anticoagulant: causes
estimation of ESR by dilution and cannot be used
Westergren method for complete blood counts
182 Essentials of Clinical Pathology

Mix to dissolve. Place 0.04 ml of this solution in a Other Tubes for Collection of Blood
bottle for 2.5 ml of blood. Anticoagulant should be dried Plain tubes (i.e. without any anticoagulant) are used for
on a warm bench or in an incubator at 37°C before use. chemistry studies after separation of serum: liver function
For routine hematological investigations, 2-3 ml of tests (total proteins, albumin, aspartate aminotransferase,
EDTA blood is required. alanine aminotransferase, bilirubin), renal function tests
Heparin (blood urea nitrogen, creatinine), calcium, lipid profile,
electrolytes, hormones, and serum osmolality.
Heparin prevents coagulation by enhancing the activity Fluoride bulb is used for collection of whole blood
of antithrombin III (AT III). AT III inhibits thrombin and for estimation of blood glucose. Addition of sodium
some other coagulation factors. It is used in the fluoride (2.5 mg/ml of blood) maintains stable glucose
proportion of 15-20 IU/ ml of blood. Sodium, lithium, or level by inhibiting glycolysis. Sodium fluoride is
ammonium salt of heparin is used. commonly used along with an anticoagulant such as
Heparin should not be used for total leukocyte count potassium oxalate or EDTA.
(since it causes leukocyte clumping) and for making of
blood films (since it imparts a blue background). SEQUENCE OF FILLING OF TUBES
It is used for osmotic fragility test (since it does not Following order of filling of tubes should be followed
alter the size of cells) and for immunophenotyping. after withdrawal of blood from the patient if multiple
Double Oxalate (Wintrobe Mixture) investigations are ordered:
1. First tube: Blood culture.
This consists of ammonium oxalate and potassium 2. Second tube: Plain tube (serum).
oxalate in 3:2 proportion. This combination is used to 3. Third tube: Tube containing anticoagulant (EDTA,
balance the swelling of red cells caused by ammonium citrate, or heparin).
oxalate and shrinkage caused by potassium oxalate. 4. Fourth tube: Tube containing additional stabilizing
Mechanism of anticoagulant action is removal of calcium. agent like fluoride.
It is used for routine hematological tests and for
estimation of erythrocyte sedimentation rate by Wintrobe USE OF PLASMA VS. SERUM
method. Plasma is the supernatant liquid obtained after centrifu-
As it causes crenation of red cells and morphologic gation of anticoagulated whole blood. Serum is the liquid
alteration in white blood cells, it cannot be used for obtained after clotting of whole blood sample collected
making of blood films. in a plain tube. Some of the differences between the two
are as follows:
Preparation
1. Plasma contains fibrinogen as well as all the other
Ammonium oxalate 1.2 gm proteins, while serum does not contain fibrinogen.
Potassium oxalate 0.8 gm 2. Plasma can be obtained immediately after sample
Distilled water upto 100 ml collection by centrifugation, while minimum of 30
Place 0.5 ml of this solution in a bottle for 5 ml of minutes are required for separation of serum from
blood. Anticoagulant should be dried in an incubator at the clotted blood.
37°C or on a warm bench before use. 3. Amount of sample is greater with plasma than with
serum for a given amount of blood.
Trisodium Citrate (3.2%) 4. Use of anticoagulant may alter the concentration of
This is the anticoagulant of choice for coagulation studies some constituents if they are to be measured like
and for estimation of erythrocyte sedimentation rate by sodium, potassium, lithium, etc.
Westergren method.
BIBLIOGRAPHY
Preparation
1. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical
Trisodium citrate 3.2 gm Hematology. 9th Ed. London: Churchill Livingstone,
Distilled water upto 100 ml 2001.
Mix well to dissolve. Store in a refrigerator at 2-8°C. 2. World Health Organization. Manual of Basic Techni-
Use 1:9 (anticoagulant: blood) proportion for coagu- ques for a Health Laboratory. 2nd Ed. Geneva: World
lation studies; for ESR, 1:4 proportion is recommended. Health Organization, 2003.
ESR should be measured within 4 hours of collection 3. World Health Organization. Use of anticoagulants in
of blood, while coagulation studies should be performed diagnostic laboratory investigations. 2002. WHO/DIL/
within 2 hours. LAB/99.1 Rev.2
18

Estimation of Hemoglobin

Hemoglobin carries oxygen from the lungs to the tissues


Box 18.2: Polycythemia
and carbon dioxide from tissues to the lungs. It is
composed of heme (iron + protoporphyrin) and globin • Primary: Increase in total red cell mass with low
polypeptide chains. Hemoglobin is not homogeneous erythropoietin level e.g. polycythemia vera
and normally different variants and derivatives exist. • Secondary: Increase in total red cell mass with
Normal hemoglobin variants are embryonic hemo- increased erythropoietin level, e.g. hypoxia
globins (Gower I, Gower II, and Portland), fetal hemo- • Relative: Decrease in plasma volume with red cell
globin (Hb F), adult hemoglobin (Hb A), and Hb A2.They mass remaining normal, e.g. dehydration.
differ from each other in the type of polypeptide chains
(Box 18.1).
2. Screening for polycythemia: Polycythemia refers to
INDICATIONS FOR HEMOGLOBIN
increased hemoglobin level above the normal range.
ESTIMATION It may be primary, secondary, or relative (Box 18.2).
1. To determine presence and severity of anemia: 3. To assess response to specific therapy in anemia.
Anemia refers to low hemoglobin concentration or 4. Estimation of red cell indices (along with packed cell
oxygen-carrying capacity of blood. Clinical signs of volume and red cell count) i.e. mean cell hemoglobin
anemia (pallor of skin, conjunctival vessels, or and mean cell hemoglobin concentration.
mucous membranes) are unreliable for diagnosis of 5. Selection of blood donors.
anemia. Anemia is best assessed by estimation of
METHODS FOR ESTIMATION OF
hemoglobin or packed cell volume (See Chapter 27:
HEMOGLOBIN
Laboratory Tests in Anemia).
There are various methods for estimation of hemoglobin.
These are:
Box 18.1: Hemoglobin 1. Colorimetric methods: In these methods, color
comparison is made between the standard and the
• Normal physiological hemoglobins: Oxyhemoglobin, test sample, either visually or by colorimetric
Reduced hemoglobin (i.e. iron of heme not associated with methods.
oxygen) • Visual methods: Tallqvist chart, Sahli’s acid
• Hemoglobin derivatives: Compounds formed from hematin method, and WHO hemoglobin color
physiological hemoglobins through the action of acids, scale.
alkalis, oxidizing/reducing agents, etc. They are identified • Photoelectric methods: Cyanmethemoglobin (hemi-
by their characteristic absorption spectra in a spectro- globincyanide) method, oxyhemoglobin method,
photometer. and alkaline hematin method.
• Hemoglobin variants: Different structural forms of 2. Gasometric method: Oxygen-carrying capacity of
hemoglobin that differ in structure of polypeptide chains. blood is measured in a Van Slyke apparatus. The
They are identified by hemoglobin electrophoresis. amount of hemoglobin is then derived from the
– Normal: HbA (α2β2), HbF (α2γ2), HbA2 (α2δ2) formula that 1 gram of hemoglobin carries 1.34 ml of
– Abnormal: HbS, HbC, HbD, HbE, etc. oxygen. However, this method measures only physio-
logically active hemoglobin, which can carry oxygen.
184 Essentials of Clinical Pathology

It does not measure carboxyhemoglobin, sulfhemo-


globin, and methemoglobin. Also, this method is
time-consuming and expensive. The result is about
2% less than other methods.
3. Chemical method: Iron-content of hemoglobin is first
estimated. Value of hemoglobin is then derived
indirectly from the formula that 100 grams of
hemoglobin contain 374 mg of iron. This method is
tedious and time-consuming.
4. Specific gravity method: A rough estimate of
hemoglobin is obtained from the specific gravity of
blood as determined from copper sulphate technique.
This method is useful in mass screening like selection
of blood donors.

Tallqvist Hemoglobin Chart


Tallqvist hemoglobin chart consists of a series of
lithographed colors said to correspond to hemoglobin
values ranging from 10 to 100%. A drop of blood obtained
by finger puncture is placed on a piece of absorbent Fig. 18.1: Sahli’s hemoglobinometer
paper. Color produced is matched against the color on
the chart and corresponding reading is taken. Although 2. Take blood sample in Sahli’s pipette exactly up to 20
this method is cheap and simple, margin of error is 20- μl mark. Blood adhering to the exterior of the pipette
50%. is wiped away using absorbent paper or gauze.
3. Add blood sample to the acid solution, mix with a
Sahli’s Acid Hematin Method
glass stirrer, and allow to stand for 10 minutes.
Principle: Blood is mixed with an acid solution so that 4. Add distilled water drop by drop till the color of the
hemoglobin is converted to brown-colored acid hematin. solution matches that of the brown glass standard.
This is then diluted with water till the brown color 5. Take the reading of the lower meniscus from the
matches that of the brown glass standard. The graduated tube in grams.
hemoglobin value is read directly from the scale.
Note:
Equipment (Fig 18.1) 1. Although the graduated tube is marked in both grams
1. Sahli’s hemoglobinometer: This consists of Sahli’s and percent figures, result should always be reported
graduated hemoglobin tube (marked in grams and in grams. This is because (a) no single hemoglobin
percent) and a comparator with a brown glass value can be considered as 100% since it varies with
standard. the age and sex of the individual and altitude, and
2. Sahli’s pipette or hemoglobin pipette (marked at 20 μl (b) hemoglobinometers of different manufacturers
or 0.02 ml). have different values as 100%, so that same sample
3. Small glass rod (stirrer). of blood will yield different results on different
4. Dropping pipette. instruments.
2. Disadvantages of Sahli’method:
Reagents • About 95% color of acid hematin is attained at the
end of 10 minutes. For maximum color develop-
1. N/10 hydrochloric acid
ment, much longer time (1 hour) is required.
2. Distilled water
• Perfect matching with the brown glass standard
Specimen: EDTA-anticoagulated venous blood or blood is not possible.
obtained by skin puncture. • Carboxyhemoglobin, methemoglobin, and sulf-
hemoglobin are not converted to acid hematin.
Method HbF is also not converted to acid hematin and
1. Place N/10 hydrochloric acid into Sahli’s graduated therefore this method is not suitable in small
hemoglobin tube up to the mark of 2 grams. infants.
Estimation of Hemoglobin 185

• Development of color is slow and acid hematin World Health Organization after extensive field trials. It
solution is not stable. is mainly intended for detection and management of
• Source of light (daylight or artificial) will influence anemia in under-resourced countries. It is especially
the visual comparison of colors. useful for screening of blood donors, for screening
• Personal error in matching brown glass standard children and women in health programs, monitoring iron
with test solution is 10%. therapy, decision- making regarding referral to a hospital,
• Color of brown glass standard fades with time. and as a point of care device.

WHO Hemoglobin Color Scale Cyanmethemoglobin (Hemiglobincyanide) Method


This method, devised by Stott and Lewis (1995), is similar This is the method of choice for estimation of hemoglobin
in principle to the now obsolete Tallqvist method. Certain and is recommended by International Committee for
technical modifications have been made to improve the Standardization in hematology. This is because (i) all
accuracy and reliability. This method is rapid, simple, forms of hemoglobin are converted to cyanmethemo-
inexpensive, and reliable within 1 gram/dl for diagnosis globin (except sulfhemoglobin), and (ii) a stable and
of anemia. The hemoglobin color scale consists of a reliable standard is available.
printed set of colors corresponding to hemoglobin values
ranging from 4-14 grams/dl (Fig. 18.2). A drop of blood Principle
is placed on a strip of chromatography paper and the Blood is mixed with a solution of potassium ferricyanide,
color developed is matched visually against the printed potassium cyanide and a non-ionic detergent (Drabkin’s
color scale. Studies have shown that efficiency is greater solution). Erythrocytes are lysed producing an evenly
than 90% in detecting anemia and 86% in classifying its distributed hemoglobin solution. Potassium ferricyanide
grade. Hemoglobin color scale has been developed by converts hemoglobin to methemoglobin, and methemo-
globin combines with potassium cyanide to form
cyanmethemoglobin (hemiglobincyanide). All forms of
hemoglobin present in blood are completely converted
to a single compound, cyanmethemoglobin. When the
reaction is completed, absorbance of the solution is
measured in a spectrophotometer at 540 nm. At this
wavelength, cyanmethemoglobin has a broad absorbance
peak. To obtain the amount of hemoglobin in the
unknown sample, its absorbance is compared with that
of the standard cyanmethemoglobin solution (the
hemoglobin concentration of which is known) by using
a formula or a previously prepared graph/table. The
reaction is linear up to 20 gm/dl.

Equipment
1. Photoelectric colorimeter or spectrophotometer
2. Sahli’s pipette marked at 20 μl (20 cmm).
3. Pipette 5 ml.

Reagents
1. Drabkin’s solution (pH 7.0-7.4):
Potassium ferricyanide 200 mg
Potassium cyanide 50 mg
Potassium dihydrogen phosphate 140 mg
Non-ionic detergent 1 ml
Distilled water to 1000 ml.
2. Cyanmethemoglobin standard solution with known
Fig. 18.2: Hemoglobin color scale hemoglobin value
186 Essentials of Clinical Pathology

Specimen: EDTA-anticoagulated venous blood or blood


obtained from skin puncture.

Method
1. In a test tube, take 5 ml of Drabkin’s solution and to
it add 20 μl of blood (1:251 dilution). Stopper the tube,
mix by inverting several times, and allow to stand
for atleast 5 minutes. This time is adequate for
conversion of hemoglobin to cyanmethemoglobin.
2. Transfer the test sample to a cuvette. Read the
absorbance in a spectrophotometer at 540 nm or in a
photoelectric colorimeter using a yellow-green filter.
Also take the absorbance of the standard solution.
Absorbance should be read against reagent blank
(Drabkin’s solution).
3. Hemoglobin value is derived from the formula given
below or from the previously prepared graph or table.

Fig. 18.3: Standard curve for determining hemoglobin


Absorbance of test sample
Hemoglobin in gm/dl = × concentration by cyanmethemoglobin method
Absorbance of standard

Dilution factor
Concentration of standard × Oxyhemoglobin Method
100
In this method, blood sample is mixed with a weak
Preparation of graph and table: If a graph or a table is ammonia solution. Absorbance of this solution is
prepared which correlates absorbance with hemoglobin measured in a spectrophotometer at 540 nm or in a
photometer using a yellow-green filter. Absorbance of
concentration, result can be obtained quickly. This is
the test sample is compared with that of the standard
particularly suitable when a large number of samples
solution.
are regularly processed on the same instrument.
This method is rapid and simple. However, no stable
Diluted cyanmethemoglobin standards are available
standard solution is available, color of oxyhemoglobin
commercially for preparation of a calibration graph.
solution rapidly fades, and hemoglobin derivatives other
Alternatively, standard cyanmethemoglobin solution is
than oxyhemoglobin are not measured.
diluted serially with Drabkin’s solution. On a linear
graph paper, hemoglobin concentration (horizontal axis) Specific Gravity Method
in each dilution is plotted against the absorbance (vertical
This is a rapid and simple method, which gives approxi-
axis). A straight line joining the points and passing
mate hemoglobin value. It is commonly used for selection
through the origin is obtained. From this graph, a table
of blood donors in blood banks.
can be prepared relating absorbance to hemoglobin A drop of blood is allowed to fall in copper sulphate
concentration (Fig. 18.3). solution of specific gravity 1.053 from a height of 1 cm.
Notes: Specific gravity of 1.053 is equivalent to hemoglobin
concentration of 12.5 grams/dl. The drop of blood gets
1. Lipemic blood (hypertriglyceridemia), high total
covered with copper proteinate and remains discrete for
leukocyte count (> 25,000/μl), or abnormal plasma
15-20 seconds. If the drop sinks within this time, its
proteins (e.g. in multiple myeloma, Waldenström’s specific gravity is higher than that of copper sulphate
macroglobulinemia) can cause erroneous results. solution (i.e. hemoglobin is >12.5 grams/dl) and
2. Cyanmethemoglobin solution is stable so that delay hemoglobin level is acceptable for donation. If it floats,
in taking the reading of absorbance does not affect hemoglobin level is unacceptable. However, specific
the result. gravity of whole blood is also affected by total leukocyte
Estimation of Hemoglobin 187

count and concentration of plasma proteins. In the • Anemia is graded as mild (hemoglobin value from
presence of leukocytosis (e.g. as in chronic myeloid lower limit of normal range to 10.0 grams/dl),
leukemia) or hypergammaglobulinemia (e.g. multiple moderate (7.0-10.0 grams/dl), and severe (< 7.0
myeloma), hemoglobin value will be misleadingly high. grams/dl).
GENERAL REMARKS
REFERENCE RANGES (WORLD HEALTH
• The cyanmethemoglobin method is the most accurate ORGANIZATION)
method for estimation of hemoglobin.
• If anemia is suspected, it is prudent to measure • Adult males: 13.0 - 17.0 gm/dl.
packed cell volume and red cell count along with • Adult females (non-pregnant): 12.0 – 15.0 gm/dl.
hemoglobin concentration. This will be useful in • Adult females (pregnant): 11.0 - 14.0 gm/dl.
assessing the correctness of hemoglobin value and in • Children, 6-12 years: 11.5 - 15.5 gm/dl.
calculation of red cell indices (for morphological • Children, 6 months to 6 years: 11.0 – 14.0 gm/dl.
classification of anemia). Packed cell volume is • Children, 2 – 6 months: 9.5 – 14.0 gm/dl.
roughly three-times the hemoglobin value. This rule, • At birth (full term): 13.6 – 19.6 gm/dl.
however, does not apply to hypochromic anemia
since red cells contain lower hemoglobin for their size. CRITICAL VALUES
• Hemoglobin level is decreased in anemia, recumbent
position (by 5-6%), excess squeezing during finger • < 7 gm/dl (severe anemia)
puncture, presence of clots in the sample, inadequate • > 20 gm/dl (hyperviscosity)
mixing of blood with anticoagulant, and “spurious”
anemia. Causes of “spurious” or “pseudo” anemia BIBLIOGRAPHY
are increased plasma volume in third trimester of 1. Cheesbrough M. District Laboratory Practice in Tropical
pregnancy, pooling of red cells in splenomegaly, fluid Countries. Part 2. Cambridge: Cambridge University
retention in congestive cardiac failure, and rise in Press, 1998.
plasma proteins in paraproteinemias. Hemoglobin 2. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical
level is increased following strenuous exercise, at high Hematology (9th Ed). London: Churchill Livingstone,
2001.
altitude, in hemoconcentration (e.g. dehydration),
3. Wallach J. Interpretation of Diagnostic Tests (7th Ed).
prolonged application of tourniquet during vene- Philadelphia: Lippincott Williams & Wilkins, 2000.
puncture, and in polycythemia. 4. World Health Organization. Manual of Basic Techni-
• On most automated hematology analyzers, hemo- ques for a Health Laboratory (2nd Ed). Geneva: World
globin is measured as cyanmethemoglobin. Health Organization, 2003.
19

Packed Cell Volume

Packed cell volume (PCV) is the volume occupied by the Internal diameter is 3 mm. It can hold about 3 ml of
red cells when a sample of anticoagulated blood is blood.
centrifuged. It indicates relative proportion of red cells 2. Pasteur pipette with a rubber bulb and a sufficient
to plasma. PCV is also called as hematocrit or erythrocyte length of capillary to reach the bottom of the Wintrobe
volume fraction. It is expressed either as a percentage of tube.
original volume of blood or as a decimal fraction. 3. Centrifuge with a speed of 2300 g.

USES OF PCV Specimen

• Detection of presence or absence of anemia or Venous blood collected in EDTA (1.5 mg EDTA for 1 ml
polycythemia of blood) or in double oxalate. Test should be performed
• Estimation of red cell indices (mean cell volume and within 6 hours of collection.
mean corpuscular hemoglobin concentration)
• Checking accuracy of hemoglobin value (Hemoglobin Method
in grams/dl × 3 = PCV). 1. Mix the anticoagulated blood sample thoroughly.
There are two methods for estimation of PCV: macro 2. Draw the blood sample in a Pasteur pipette and
method (Wintrobe method) and micro method (micro- introduce the pipette up to the bottom of the Wintrobe
hematocrit method). Micro method is preferred because tube. Fill the tube from the bottom exactly up to the
it is rapid, convenient, requires only a small amount of 100 mark. During filling, tip of the pipette is raised,
blood, capillary blood from skin puncture can be used, but should remain under the rising meniscus to avoid
and a large number of samples can be tested at one time. foaming.
This method is also more accurate as plasma trapping in 3. Centrifuge the sample at 2300 g for 30 min (To
red cell column is less. counterbalance a second Wintrobe tube filled with
blood from another patient or water should be placed
MACRO METHOD (WINTROBE METHOD) in the centrifuge).
4. Take the reading of the length of the column of red
Principle
cells. Hematocrit can be expressed either as a
Anticoagulated whole blood is centrifuged in a Wintrobe percentage or as a fraction of the total volume of blood
tube to completely pack the red cells. The volume of sample.
packed red cells is read directly from the tube.
An advantage with this method is that before Significance
performing PCV, test for erythrocyte sedimentation rate In anemia, PCV is below the lower level of normal range.
can be set up. PCV is raised in dehydration, shock, burns, and
polycythemia.
Equipment
After centrifugation of anticoagulated whole blood,
1. Wintrobe tube: This tube is about 110 mm in length three zones can be distinguished in the Wintrobe tube
and has 100 markings, each at the interval of 1 mm. from above downwards- plasma, buffy coat layer (a small
Packed Cell Volume 189

Fig. 19.1: Anticoagulated blood-filled Wintrobe hematocrit tubes Fig. 19.2: Preparation of smear from buffy coat
after centrifugation, showing normal PCV, low PCV (anemia),
and thick buffy coat layer

greyish layer of white cells and platelets, about 1 mm


thick), and packed red cells (Fig. 19.1). Normal plasma is
straw-colored. It is colorless in iron deficiency anemia,
pink in the presence of hemolysis or hemoglobinemia,
and yellow if serum bilirubin is raised (jaundice). In
hypertriglyceridemia, plasma appears milky. Increased
thickness of buffy coat layer occurs if white cells or
platelets are increased in number (e.g. in leukocytosis,
thrombocytosis, or leukemia). Smears can be made from
the buffy coat layer (Fig. 19.2) for demonstration of lupus
erythematosus (LE) cells (Fig. 19.3), malaria parasites, or
immature cells.

MICRO METHOD Fig. 19.3: Lupus erythematosus or LE cell (red arrow) in buffy
coat smear. LE cell is a neutrophil with a homogeneous red
Principle purple inclusion distending the cytoplasm. It is seen in collagen
disorders, drug reactions, chronic hepatitis, and serum sickness
Anticoagulated whole blood is centrifuged in a capillary
tube of uniform bore to pack the red cells. Centrifugation
is done in a special microhematocrit centrifuge till
packing of red cells is as complete as possible. The
reading (length of packed red cells and total length of
the column) is taken using a microhematocrit reader, a
ruler, or arithmetic graph paper (Fig. 19.4).

Equipment
1. Microhematocrit centrifuge: It should provide relative
centrifugal force of 12000 g for 5 minutes.
2. Capillary hematocrit tubes: These are disposable glass Fig. 19.4: Microhematocrit capillary tubes following
tubes 75 mm in length and 1 mm in internal diameter. centrifugation in a microhematocrit centrifuge
190 Essentials of Clinical Pathology

They are of two types: plain (containing no anticoagu- GENERAL NOTES


lant) and heparinised (coated with a dried film of 2
1. Prolonged application of tourniquet during
units of heparin). For plain tubes, anticoagulated
venepuncture causes hemoconcentration and rise
venous blood is needed. Heparinised tubes are used
for blood obtained from skin puncture. in hematocrit.
3. Tube sealant like plastic sealant or modeling clay; if 2. Excess squeezing of the finger during skin
not available, a spirit lamp for heat sealing. puncture dilutes the sample with tissue fluid and
4. Microhematocrit reader; if not available, a ruler or lowers the hematocrit.
arithmetic graph paper. 3. Correct proportion of blood with anticoagulant
should be used. Excess EDTA causes shrinkage
Specimen of red cells and falsely lowers the hematocrit.
4. Inadequate mixing of blood with anticoagulant,
Venous blood collected in EDTA (dipotassium salt) for
and inadequate mixing of blood before testing can
plain tubes or blood from skin puncture collected directly
cause false results.
in heparinised tubes. Venous blood should be collected
5. Low hematocrit can result if there are clots in the
with minimal stasis to avoid hemoconcentration and
sample.
false rise in PCV.
6. Centrifugation at lower speed and for less time
Method falsely increases PCV.
7. A small amount of plasma is trapped in the lower
1. Fill the capillary tube by applying its tip to the blood
part of the red cell column which is usually
(either from skin puncture or anticoagulated venous
insignificant. Increased amount of plasma is
blood, depending on the type of tube used). About
trapped in microcytosis, macrocytosis, sphero-
2/3rds to 3/4ths length of the capillary tube should
cytosis, and sickle cell anemia, which cause an
be filled with blood.
2. Seal the other end of the capillary tube (which was artifactual rise in hematocrit. Larger volume of
not in contact with blood) with a plastic sealant. If it plasma is trapped in Wintrobe tube than in
is not available, heat-seal the tube using a spirit lamp. capillary tube.
3. The filled tubes are placed in the radial grooves of 8. As PCV requires whole blood sample, it is affected
the centrifuge with the sealed ends toward the outer by plasma volume (e.g. PCV is higher in dehy-
rim gasket. Counterbalance by placing the tubes in dration, and lower in fluid overload).
the grooves opposite to each other. 9. Expression of PCV: Occasionally, PCV is
4. Centrifuge at relative centrifugal force 12000 g for 5 expressed as a percentage. In SI units, PCV is
minutes to completely pack the red cells. expressed as a volume fraction. Conversion factor
5. Immediately remove the tubes from the centrifuge from conventional to SI units is 0.1 and from SI to
and stand them upright. The tube will show three conventional units is 100.
layers from top to bottom: column of plasma, thin 10. Rules of 3 and 9: These rules of thumb are
layer of buffy coat, and column of red cells.
commonly used to check the accuracy of results
6. With the microhematocrit reader, hematocrit is
and are applicable only if red cells are of normal
directly read from the scale. If hematocrit reader is
size and shape.
not available, the tube is held against a ruler and the
hematocrit is obtained by the following formula:
Hemoglobin (gm/dl) × 3 = PCV

Length of red cell column in mm Red cell count (million/cmm) × 9 = PCV


Length of total column in mm
11. Automated hematocrit: In automated hematology
To obtain PCV, the above result is multiplied by 100. analyzers, hematocrit is obtained by multiplying
Packed Cell Volume 191

red cell count (in millions/cmm) by mean cell CRITICAL VALUES


volume (in femtoliters). • Packed cell volume: < 20% or > 60%
REFERENCE RANGES BIBLIOGRAPHY
• Adult males: 40-50% 1. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical
• Adult females (nonpregnant): 38-45% Hematology (9th Ed). London: Churchill Livingstone,
• Adult females (pregnant): 36-42% 2001.
2. Wallach J. Interpretation of Diagnostic tests (7th Ed).
• Children 6 to 12 years: 37-46% Philadelphia: Lippincott Williams and Wilkins, 2000.
• Children 6 months to 6 years: 36-42% 3. World Health Organization. Manual of Basic Techni-
• Infants 2 to 6 months: 32-42% ques for a Health Laboratory (2nd Ed). Geneva. World
• Newborns: 44-60% Health Organization, 2003.
20

Total Leukocyte Count

Total leukocyte count (TLC) refers to the number of white


blood cells in 1 μl of blood (or in 1 liter of blood if the
result is expressed in SI units). There are two methods
for estimation of TLC:
• Manual or microscopic method
• Automated method (See Chapter 32: Automation in
Hematology).
A differential leukocyte count should always be
performed along with TLC to obtain the absolute cell
counts.
The purpose of carrying out TLC is to detect increase
or decrease in the total number of white cells in blood,
i.e. leukocytosis or leukopenia respectively. TLC is carried
out in the investigation of infections, any fever,
hematologic disorders, malignancy, and for follow-up
of chemotherapy or radiotherapy.
Fig. 20.1: Neubauer counting chamber: (A) Surface view,
MANUAL METHOD (B) Side view

Principle
0.1 ml. In the improved Neubauer chamber, the
A sample of whole blood is mixed with a diluent, which
central large square is divided into 25 squares, each
lyses red cells and stains nuclei of white blood cells. White
of which is further subdivided into 16 small squares.
blood cells are counted in a hemocytometer counting
A group of 16 small squares is separated by closely
chamber under the microscope and the result is
ruled triple lines (Figs 20.2 and 20.3). Metallized
expressed as total number of leukocytes per μl of blood
surface makes background rulings and cells easily
or per liter of blood.
visible. The 4 large corner squares are used for
Equipment counting leukocytes, while the central large square
is used for counting platelets and red blood cells.
1. Hemocytometer or counting chamber with coverglass: The Only special coverglass, which is intended for
recommended hemocytometer is one with improved use with hemocytometer, should be used. It should
Neubauer rulings and metallized surface. There are be thick and optically flat. When the special
two ruled areas on the surface of the chamber coverglass is placed on the surface of the chamber, a
(Fig. 20.1). Each ruled area is 3 mm × 3 mm in size volumetric chamber with constant depth and volume
and consists of 9 large squares with each large square throughout its entire area is formed. Ordinary cover
measuring 1 mm × 1 mm. When the special thick slips should never be employed since they do not
coverglass is placed over the ruled area, the volume provide constant depth to the underlying chamber
occupied by the diluted blood in each large square is due to bowing.
Total Leukocyte Count 193

Fig. 20.2: Neubauer counting chamber showing areas for Fig. 20.4: (A) WBC pipette and (B) Sahli’s pipette
counting WBCs (W) and red blood cells and platelets (R) calibrated to deliver 20 μl

3. Graduated pipette, 1 ml.


4. Pasteur pipette
5. Test tube (75 × 12 mm).

Reagent
WBC diluting fluid (Turk’s fluid) consists of a weak acid
solution (which hemolyzes red cells) and gentian violet
(which stains leucocyte nuclei deep violet). Diluting fluid
also suspends and disperses the cells and facilitates
counting. Its composition is as follows:
• Acetic acid, glacial 2 ml
Fig. 20.3: (A) Counting area for WBCs and (B) counting • Gentian violet, 1% aqueous 1 ml
area for RBCs and platelets in Neubauer counting chamber • Distilled water to make 100 ml

Specimen
When the special cover glass is placed over the
ruled area of the chamber and pressed, Newton’s EDTA anticoagulated venous blood or blood obtained
rings (colored refraction or rainbow colored rings) by skin puncture is used. (Heparin should not be used
appear between the two glass surfaces; their since it causes leukocyte clumping). While collecting
formation indicates the correct placement of the cover capillary blood from the finger, excess squeezing should
be avoided so as not to dilute blood with tissue fluid.
glass.
2. Pipette calibrated to deliver 20 μl (0.02 ml, 20 cmm): WBC
Method
bulb pipettes (Fig. 20.4), which have a bulb for
dilution and mixing (Thoma pipettes) are no longer 1. Dilution of blood: Take 0.38 ml of diluting fluid in a
recommended. This is because blood and diluting test tube. To this, add exactly 20 μl of blood and mix.
fluid cannot be mixed adequately inside the bulb of This produces 1:20 dilution. Alternatively, 0.1 ml of
the pipette. Bulb pipettes are also difficult to calibrate, blood can be added to 1.9 ml of diluting fluid to get
costly, and charging of counting chamber is difficult. the same dilution.
Tips of pipettes often chip easily and unnecessarily 2. Charging the counting chamber: Place a coverglass
small volume of blood needs to be used. over the hemocytometer. Draw some of the diluted
194 Essentials of Clinical Pathology

blood in a Pasteur pipette. Holding the Pasteur pipette conventional to SI units is 0.001 and SI to conventional
at an angle of 45° and placing its tip between the units is 1000.
coverglass and the chamber, fill one of the ruled areas
Correction of TLC for nucleated red cells: The diluting
of the hemocytometer with the sample. The sample
fluid does not lyse nucleated red cells or erythroblasts.
should cover the entire ruled area, should not contain
Therefore, they are counted as leukocytes in hemo-
air bubbles, and should not flow into the side
cytometer. If erythroblasts are markedly increased in the
channels. Allow 2 minutes for settling of cells.
blood sample, overestimation of TLC can occur. To avoid
3. Counting the cells: Place the charged hemocytometer
this if erythroblasts are greater than 10 per 100 leukocytes
on the microscope stage. With the illumination
as seen on blood film, TLC should be corrected for
reduced to give sufficient contrast, bring the rulings
nucleated red cells by the following formula:
and the white cells under the focus of the low power
objective (× 10). White cells appear as small black dots.
Count the number of white cells in four large corner TLC/μl × 100
squares. (To reduce the error of distribution, counting Corrected TLC/μl = ————————————
Nucleated red cells per
of cells in all the nine squares is preferable). To correct 100 WBCs + 100
for the random distribution of cells lying on the
margins of the square, cells which are touching the
Sources of error in manual blood cell count: There are
left hand lines or upper lines of the square are
two main errors: technical and inherent.
included in the count, while cells touching the lower
and right margins are excluded. Technical Errors

4. Calculation of TLC: Errors in blood collection:


• Prolonged, tight application of a tourniquet leads to
(1) TLC/μl = (Number of WBCs counted × venous stasis and false elevation of cell count.
Correction for dilution × Correction for volume) • Excessive squeezing of finger puncture results in
——————————————————————— dilution of blood with tissue fluid.
(Number of large squares counted)
• Inadquate mixing of blood with anticoagulant leads
Number of WBCs counted × 20 × 10 to formation of clots in blood sample and falsely low
= ————————————————— count.
4
• Non-mixing of anticoagulated venous blood imme-
= Number of WBCs counted × 50
diately before testing will cause falsely low count.

(2) TLC/L = Number of WBCs counted × 50 × 106 (106 is Errors in pipetting


the correction factor to convert count in 1 μl to count in 1 • Use of wet, chipped, or dirty pipettes
• Use of improperly calibrated pipettes
liter).
• Not wiping blood adherent to outside of pipette
Example: If 200 WBCs are counted in 4 large squares, • Using bulb pipettes that are difficult to calibrate and
TLC/μl will be 10,000/μl and TLC/liter will be 10.0 × which break off easily.
109/liter. Errors in filling of chamber
If TLC is more than 50,000/ml, then dilution of blood • Incorrect filling of chamber, spillage into moats
should be increased to 1:40 to increase the accuracy of • Not allowing 2 minutes for cells to settle.
the result. • Drying of edges of preparation
If TLC is less than 2,000/ml then lesser dilution • Not using specified coverglass
should be used. • Chamber containing dirt or air bubble.

Expression of TLC: Conventionally, TLC is expressed as Inherent Error (Statistical Error)


cells/μl or cells/cmm or cells/mm3. In SI units, TLC is In spite of the best technique, an error still occurs due to
expressed as cells × 109/liter. Conversion factors for random distribution of cells in the chamber. This results
Total Leukocyte Count 195

in variation in cell number in different areas of the same CRITICAL VALUES


size. This distribution follows Poisson’s law. More Total leukocyte count< 2000/μl or > 50000/μl.
number of cells should be counted to reduce this error.
n
This variance is calculated from the formula  100% , BIBLIOGRAPHY
n
where n is the total number of cells counted. If 100 cells are
1. Cheesbrough M. District laboratory practice in tropical
counted, then variance will be 10%. In 95% of cases, the
countries. Part 1 and Part 2. Cambridge. Cambridge
range would be n ± 2σ i.e. 100 –2(10) to 100 + 2(10), i.e. 80 University Press, 1998.
to 120. If 200 cells are counted, the error is reduced to 2. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical
about 7%. Haematology (9th Ed). London. Churchill Livingstone,
2001.
REFERENCE RANGES 3. The Expert Panel on Cytometry of the International
• Adults: 4000-11,000/μl Council for Standardization in Haematology: Recom-
mended methods for the visual determination of white
• At birth: 10,000-26000/μl
blood cell count and platelet count. World Health
• 1 year: 6,000-16,000/μl
Organization. WHO/DIL/00.3. 2000.
• 6-12 years: 5,000-13,000/μl 4. World Health Organization. Manual of Basic Techni-
• Pregnancy: up to 15,000/μl ques for a Health Laboratory (2nd Ed). Geneva: World
Health Organization, 2003.
21

Reticulocyte Count

Reticulocytes are young or juvenile red cells released 4. To assess response to erythropoietin therapy in
from the bone marrow into the bloodstream and that anemia of chronic renal failure.
contain remnants of ribonucleic acid (RNA) and 5. To follow the course of bone marrow transplantation
ribosomes but no nucleus. After staining with a for engraftment
supravital dye such as new methylene blue, RNA 6. To assess recovery from myelosuppressive therapy
appears as blue precipitating granules or filaments within 7. To assess anemia in neonate
the red cells. Following supravital staining, any non-
nucleated red cell containing 2 or more granules of blue- PRINCIPLE
stained material is considered as a reticulocyte (The
A few drops of blood (collected in EDTA) are incubated
College of American Pathology). Supravital staining
with new methylene blue solution which stains granules
refers to staining of cells in a living state before they are
of RNA in red cells. A thin smear is prepared on a glass
killed by fixation or drying or with passage of time.
slide from the mixture and reticulocytes are counted
Reticulocyte count is performed by manual method.
under the microscope. Number of reticulocytes is
Recently methods based on flow cytometry have been
expressed as a percentage of red cells.
introduced which are rapid and more precise.
REAGENT
Reticulocyte count is performed to assess erythropoietic
activity of the bone marrow New methylene blue solution is prepared as follows:
New methylene blue 1.0 gm
USES Sodium citrate 0.6 gm
Sodium chloride 0.7 gm
1. As one of the baseline studies in anemia with no Distilled water 100 ml
obvious cause Reagent should be kept stored in a refrigerator at
2. To diagnose anemia due to ineffective erythropoiesis 2-6°C and filtered before use.
(premature destruction of red cell precursors in bone Suitable alternatives to new methylene blue are
marrow seen in megaloblastic anemia and thalasse- brilliant cresyl blue and azure B.
mia) or due to decreased production of red cells: In
hypoplastic anemia or in ineffective erythropoiesis, SPECIMEN
reticulocyte count is low as compared to the degree Capillary blood or EDTA-anticoagulated venous blood
of anemia. Increased erythropoiesis (e.g. in hemolytic can be used.
anemia, blood loss, or specific treatment of nutritional
anemia) is associated with increased reticulocyte METHOD
count. Thus reticulocyte count is used to differentiate 1. Take 2-3 drops of filtered new methylene blue
hypoproliferative anemia from hyperproliferative solution in a 12 × 75 mm test tube.
anemia. 2. Add equal amount of blood and mix well.
3. To assess response to specific therapy in iron 3. Keep the mixture at room temperature or at 37°C for
deficiency and megaloblastic anemias. 15 minutes.
Reticulocyte Count 197

Fig. 21.1: Reticulocytes stained with a supravital stain

Fig. 21.2: Miller ocular disk to facilitate counting of reticulocytes.


4. After gentle mixing, place a small drop from the The Miller ocular disk contains two squares: A and B. Square
mixture on a glass slide, prepare a thin smear, and A is 9 times larger than square B. Square B is used for counting
allow to dry in the air. red cells, while reticulocytes are counted in square A. Only
5. Examine under the microscope using oil-immersion the reticulocytes are counted in 20 large (i.e. A) squares, while
red cells are counted in 20 small (i.e. B) squares. Reticulocyte
objective. Mature red cells stain pale green-blue.
count is obtained from the formula
Reticulocytes show deep blue precipitates of fine
granules and filaments in the form of a network Total reticulocytes counted in square A
——————————————————— × 100
(reticulum) (Fig. 21.1). Most immature reticulocytes Total red cells counted in square B × 9
show a large amount of precipitated material in the
form of a mass, while the most mature reticulocytes
show only a few granules or strands. Any non- 2. Absolute reticulocyte count
nucleated red cell is considered as a reticulocyte if it = Reticulocyte percentage × Red cell count
contains 2 or more blue-stained particles of ribosomal Normal: 50,000 to 85,000/cmm
RNA.
6. Count 1000 red cells and note the number of red cells 3. Corrected reticulocyte count (Reticulocyte index)
that are reticulocytes. Counting error is minimized if PCV of patient
size of the microscopic field is reduced. This is = Reticulocyte percentage × ————————
Normal PCV
achieved by using a Miller ocular disk inserted in the
eyepiece (Fig. 21.2); it divides the field into two Corrected reticulocyte count > 2% indicates reticulo-
squares (one nine times larger in size than the other). cyte release appropriate for the degree of anemia. If <
Reticulocytes are counted in both the squares and the 2%, reticulocyte release is inappropriate.
red cells are counted in the smaller square.
(4) Reticulocyte maturation production index
REPORTING THE RESULT Corrected reticulocyte count
= ———————————————
1. Reticulocyte percentage: The most common method Estimated maturation time in days
of reporting is reticulocyte percentage which is
calculated from the following formula: Notes:
1. Stages of maturation of reticulocytes (Heilmeyer and
Reticulocyte Westhaeuser, 1932) (Fig. 21.3):
Number of reticulocytes counted
percentage = ———————————————— × 100 • Stage 0: Late or orthochromatic normoblast
Number of red cells counted (Nucleated red cell); strong staining for reticulum
(This cell type should not be included under
Reference range is 0.5%-2.5% in adults and children. reticulocytes as it is not a stage of reticulocyte
Reticulocyte count is higher in newborns. maturation)
198 Essentials of Clinical Pathology

follows: hematocrit 45%: maturation time 1 day, 35%:


1.5 days, 25%: 2 days, and 15%: 2.5 days. There is
evidence that maturation of reticulocytes also occurs
in vitro following blood collection and therefore
specimen should be processed for reticulocyte
counting as soon as possible (within 4 hours of
collection).
3. An example illustrating importance of absolute
reticulocytes count and corrected reticulocyte count:
A 30-year-old male patient with anemia has PCV 15%,
red cell count 1.2 million count/cmm, and reticulo-
cyte count 3%. Reticulocyte count appears to be
increased (normal: 0.5-2.5%) indicating adequate
Fig. 21.3: Stages of reticulocyte maturation response to anemia. However, value of absolute
reticulocyte count (0.03 × 1200000) is 36,000 and
15
corrected reticulocyte count is 3 × or 1%; both of
45
which are low as compared to the degree of anemia.
• Stage I: Dense cohesive reticulum in non-nucleated
Thus the patient has diminished production of red
red cell (0.1% of reticulocyte count in normal
cells in marrow (hypoproliferative anemia).
individuals)
4. Causes of increased reticulocyte count (reticulo-
• Stage II: Extensive network of loose reticulum
cytosis):
(0.7% of reticulocyte count in normal individuals)
• Hemolytic anemias
• Stage III: Small reticulum along with scattered
• Blood loss
granules (32% of reticulocyte count in normal
• Following specific therapy of nutritional anemia
individuals)
(like iron in iron deficiency anemia, folate in folate
• Stage IV: Scattered granules (61% of reticulocyte
count in normal individuals). deficiency anemia, Vit B 12 in B 12 deficiency
2. Time required for maturation of reticulocytes (i.e. anemia)
from extrusion of nucleus till no granules are seen in • Hemoglobinopathies, e.g. sickle cell anemia.
the cytoplasm) is 4 to 4.5 days. Till the extrusion of 5. Causes of decreased reticulocyte count (reticulo-
the nucleus, 97% of hemoglobin is synthesized; cytopenia):
reticulocytes synthesize remaining 3%. Reticulocytes • Aplastic anemia and pure red cell aplasia
are produced in the bone marrow where they spend • Bone marrow infiltration (leukemia, lymphoma,
about 3 to 3.5 days for maturation. In circulation, they myelofibrosis, metastatic malignancy)
spend 1 more day before maturing into red cells. In • Renal disease
hemolytic anemia and following acute blood loss, • Anemia of chronic disease
reticulocytes are released prematurely in circulation • Alcoholism
where they require more time (about 2-3 days) for • Myxedema
maturation. Such prematurely released reticulocytes • Ineffective erythropoiesis: Megaloblastic anemia,
are larger than reticulocytes released under basal sideroblastic anemia, thalassemia, myelodysplasia
conditions. The maturation time is dependent upon • Following blood transfusion.
the hematocrit of the patient. Therefore, to avoid 6. Classification of anemia according to reticulocyte
overestimation of actual red cell production, count: Normocytic normochromic anemias are
reticulocyte index is calculated. In a patient with classified into two types based on reticulocyte count:
anemia, if reticulocyte index fails to rise 2-3 times • Normocytic normochromic anemia with increa-
above the normal value, then decreased production sed reticulocyte count:
of red cells in the marrow should be suspected. –Recent blood loss
Maturation time according to the hematocrit is as –Hemolysis
Reticulocyte Count 199

• Normocytic normochromic anemia with low • HbH inclusions: These are round, golf ball like
reticulocyte count: inclusion bodies seen in α-thalassemias. They are
– Aplastic anemia seen in most of the red cells while reticulocytes
– Chronic renal failure are seen in only a few cells.
– Anemia of chronic disease
• Granules of new methylene blue superimposed on
– Anemia due to ineffective erythropoiesis
red cells, if stain is not filtered before use.
7. Reticulocytes should be distinguished from (Fig. 21.4):
• Heinz bodies: These are precipitated globin chains • Howell-Jolly bodies: These are nuclear remnants in
attached to the red cell membrane. They stain light red cells seen in certain anemias and following
blue and are seen in glucose-6-phosphate dehy- splenectomy. As supravital dyes stain both DNA
drogenase deficiency following exposure to and RNA, these structures are also stained.
oxidant stress. 8. The polychromatic red cells on Romanowsky-stained
• Pappenheimer bodies: These are iron-containing blood smears represent reticulocytes. However, only
granules which appear as one or more small dots stages 1, 2, and 3 cause polychromasia, while stage 4
in red cells. They give positive Prussian blue reticulocytes having small amount of RNA do not
reaction. They are confused with reticulocytes in
cause polychromasia. Therefore, counting polychro-
Romanowsky-counterstained preparations.
matic cells cannot be used as a substitute for
reticulocyte count.

REFERENCE RANGES

• Reticulocyte percentage: 0.5-2.5%


• Absolute reticulocyte count: 50,000-85,000/cmm

BIBLIOGRAPHY

1. Pierre RV. Reticulocytes: Their usefulness and


measurement in peripheral blood. Clin Lab Med
2002;22:63-79.
2. The Expert Panel on Cytometry of the International
Council for Standardization in Hematology: ICSH
guidelines for reticulocyte counting by microscopy on
Fig. 21.4: Differentiation of reticulocytes from Howell-Jolly supravitally stained preparations. World Health
bodies, Heinz bodies, and HbH inclusions Organization. WHO/LBS/92.3 1992.
22

Blood Smear

A blood smear or film is a specimen for microscopic blood is used, smear should be prepared and stained
examination prepared by spreading a drop of blood within 2 hours of blood collection. If venous blood
across a glass slide followed by staining with one of the collected in a syringe is used, the last drop of blood
Romanowsky's stains. in the needle after withdrawing (or first drop while
dispensing) should be used.
USES 2. A 'spreader' slide is placed at an angle of 30° in front
of the drop and then drawn back to touch the drop of
1. Blood smear is helpful in suggesting the cause of
blood. Blood spreads across the line of contact of two
anemia or thrombocytopenia, identifying and typing
slides.
of leukemia, and in diagnosing hemoparasitic
3. Smear is made by smooth, forward movement of the
infections (malaria, filaria, and trypanosomiasis). It
'spreader' along the slide. The whole drop should be
is also helpful in the management of these conditions.
used up 1 cm before the end of the slide. The length
2. To monitor the effect of chemotherapy and radio-
of the smear should be about 3 cm (Fig. 22.1). The
therapy on bone marrow.
'spreader' should not be raised above the slide surface
3. To provide direction for further investigations that
till whole drop of blood is spread out.
will help in arriving at the correct diagnosis (e.g. in
infections, drug toxicity, etc.).
Blood smear examination is therefore indicated in
clinically suspected cases of anemia, thrombocytopenia,
hematological malignancies (leukemia, lymphoma,
multiple myeloma), disseminated intravascular coagu-
lation, parasitic infections (like malaria or filaria),
infectious mononucleosis, and various inflammatory, or
malignant diseases.

PREPARATION OF BLOOD SMEAR


(WEDGE METHOD)
1. A small drop of blood (2-3 mm in diameter) is placed
in the center line about 1 cm away from one end of a
glass slide (typical size of slide is 75 × 25 mm;
thickness about 1mm) with a wooden stick or glass
capillary. Slide should be clean, dry, and grease-free.
Blood sample may be venous (anticoagulated with
EDTA) or capillary (finger prick). Better blood cell
morphology is obtained if smear is made directly
from a skin puncture. If EDTA-anticoagulated venous Fig. 22.1: Preparation of blood smear
Blood Smear 201

4. Smear is rapidly dried by waving it in the air or


keeping it under an electric fan. Slow drying causes
shrinkage artifact of red cells.
5. Patient's name or laboratory number and date are
written (with a lead pencil, a permanent marker pen,
or a diamond pencil) on the thicker end of the smear.
6. The smear is fixed immediately with absolute methyl
alcohol (which should be moisture- and acetone-free)
for 2-3 minutes in a covered jar (Absolute ethyl
alcohol can also be used, but not methylated spirit as
it contains water). Aim of fixation is to prevent
washing off of the smear from the slide. Following
this, color of the smear becomes light brown. This Fig. 22.2: Blood smears that are unsatisfactory for
evaluation
fixation is desirable even when Leishman stain is used
which contains methyl alcohol. This is because
Leishman stain may have absorbed moisture leading In patients with polycythemia, a thinner smear is
to poor fixation. If methanol is contaminated with obtained by decreasing the 'spreader' angle and the
water, sharpness of cell morphology is lost and there speed of spreading.
is vacuolation of red cells. Methanol should be 4. Anticoagulant used may be EDTA (dipotassium salt)
acetone-free since acetone washes out nuclear stain. or sodium citrate. Heparin should not be used as an
(In many laboratories, slide is stained immediately anticoagulant for making blood films since it causes
after air-drying without prior fixation, and the results platelet clumping and imparts a blue background to
are satisfactory; however, if delay of >4 hours is the film.
anticipated between air-drying and staining, the slide 5. It is recommended to stain blood films in reagent
should be fixed. If not, a background gray-blue filled Coplin jars (rather than covering them with the
staining of plasma occurs). staining solution) to avoid formation of stain
precipitates due to evaporation.
Notes 6. A drawback of this method is uneven distribution of
1. Making a 'spreader' slide—a glass slide with leukocytes (i.e. monocytes, neutrophils, and abnormal
absolutely smooth edges should be selected and one cells are pushed towards the extreme tail end of the
or both corners at one end of the slide should be smear) and distortion of red cell morphology at the
broken off. The 'spreader' slide should be narrower edges.
(width of about 15 mm) so that edges of the smear 7. Blood smear is covered with a coverslip and mounted
can be examined microscopically (Fig. 22.1). The in a mounting medium (e.g. DPX) for protection
'spreader' slide should be discarded after use. If the against mechanical damage and deterioration of
same is to be reused, its edge should be thoroughly staining with time on exposure to air.
cleaned and dried (otherwise carryover of cells or 8. Cleaning of slides: (A) New slides: If new slides are
parasites can occur). not clean and grease-free, they are left overnight in a
2. A well-spread blood smear (a) is tongue-shaped with detergent solution, washed in running tap water,
a smooth tail, (b) does not cover the entire area of the rinsed in distilled water, and wiped dry with a clean
slide, (c) has both thick and thin areas with gradual cloth. Before use, they are wiped with 95% methyl
transition, and (d) does not contain any lines or holes. alcohol, dried, and then kept covered to protect from
Smears unsatisfactory for examination are shown in dust. (B) Used slides: The used slides are soaked in a
Figure 22.2. detergent solution at 60°C for 20 minutes, washed in
3. By changing the angle of the 'spreader' and its speed, running tap water, rinsed in distilled water, and then
thickness of the blood smear can be controlled. In wiped dry. Before use, they are wiped with 95%
patients with anemia, a thicker smear can be obtained methyl alcohol, dried, and then kept covered to
by increasing the angle and the speed of spreading. protect from dust.
202 Essentials of Clinical Pathology

STAINING OF BLOOD SMEAR A well-stained smear is pink in color in thinner


Blood smears are routinely stained by one of the portion and purple-blue in thicker portion. Excess blue
Romanowsky stains (Box 22.1). Romanowsky stains coloration can be due to: (i) excessively thick smear, (ii)
consist of a combination of acidic and basic dyes and low concentration of eosin, (iii) impure dyes, (iv) too long
after staining various intermediate shades are obtained staining time, (v) inadequate washing, or (vi) excessive
between the two polar (red and blue) stains. Romanow- alkaline pH of stain, buffer, or water. Excess red
sky stains include May-Grunwald-Giemsa, Jenner, coloration can be due to: (i) impure dyes or incorrect
Wright's, Leishman's, and Field's stains. Staining proportion of dyes, (ii) excessive acid pH of stain, buffer,
properties of the Romanowsky stains are dependent on or water (as the red cells take up more acid dye i.e. eosin),
two synthetic dyes: methylene blue and eosin. Inter- (iii) too short staining time, or (iv) excessive washing. If
national Committee for Standardization in Haematology there are granules of stain precipitate (masses of small
has recommended a highly purified standardized stain, black dots) on smear, stain needs to be filtered.
which contains azure B and eosin Y; it, however, is very Method of Leishman staining is given below:
expensive. Romanowsky stains are insoluble in water but
soluble in methyl alcohol. Methyl alcohol acts as a solvent Reagents
as well as a fixative. Staining reaction is pH-dependent.
These stains have a tendency towards precipitation and 1. Leishman stain: William Boog Leishman, a British
should be filtered before use. pathologist, modified the original Romanowsky
Methylene blue and azure B are basic (cationic) dyes method and devised a stain which is widely known
and have affinity for acidic components of the cells (like as Leishman's stain. This consists of methylene blue
nucleic acids or basophil granules) and impart purple- and eosin dissolved in absolute methyl alcohol.
violet color to the nuclear chromatin, dark blue-violet Commercially available Leishman stain powder (0.6
color to the basophil granules, and deep blue color to the gram) is mixed with water-free absolute methyl
cytoplasm of lymphocytes. Eosin is an acidic (anionic) alcohol (400 ml). Prepared stain should be kept tightly
dye and has affinity for basic components like hemo- stoppered in a brown bottle and stored in a cool, dark
globin (stained pink-red), and granules in eosinophils
place at room temperature. Exposure to direct
(stained orange-red). Neutrophil granules are slightly
sunlight causes deterioration of the stain. After
basic and stain violet-pink or lilac.
preparation, stain should be kept for 3-5 days before
Romanowsky stains impart more colours than just
using since it improves the quality of the stain.
blue (from methylene blue or azure B) and red-orange
2. Buffered water (pH 6.8).
(from eosin Y). Usefulness of the Romanowsky stains lies
in their ability to differentially stain leucocyte granules.
Method

1. Air-dry the smear and fix with methanol for 2-3


Box 22.1: Romanowsky stains minutes.
Two main components of all Romanowsky stains are an acidic 2. Cover the smear with Leishman stain for 2 minutes.
dye (eosin Y) and a basic dye (oxidized methylene blue). 3. After 2 minutes, add twice the volume of buffered
• Basic or cationic dye: It is positively charged and binds to water and leave for 5-7 minutes. A scum of metallic
anionic sites and imparts blue-gray color to nucleic acids, sheen forms on the surface.
nucleoproteins, and granules of basophils. Examples: 4. Wash the stain away in a stream of buffered water.
methylene blue, azure B. Tap water can also be used for washing if it is not
• Acidic or anionic dye: It is negatively charged and binds
highly alkaline or highly acid.
to cationic sites and imparts orange-red color to
5. Wipe the back of the slide clean and set it upright in
hemoglobin and eosinophil granules. Example: eosin Y.
the draining rack to dry.
Examples of Romanowsky stains: May-Grunwald-Giemsa,
6. Mount the slide in a suitable mounting medium (e.g.
Jenner’s, Leishman’s, Wright’s, Field’s.
DPX) with a clean and dry 25 × 25 mm coverslip.
Blood Smear 203

A well-stained smear shows following features


(Fig. 22.3):
• Red cells: pink-red or deep pink
• Polychromatic cells (Reticulocytes): Gray-blue
• Neutrophils: Pale pink cytoplasm; mauve-purple
granules
• Eosinophils: Pale-pink cytoplasm; orange-red
granules
• Basophils: Blue cytoplasm; dark blue-violet
granules
• Monocytes: Gray-blue cytoplasm; fine reddish
(azurophil) granules
• Small lymphocytes: Dark blue cytoplasm
• Platelets: Purple
• Nuclei of all cells: Purple-violet

EXAMINATION OF BLOOD SMEAR Fig. 22.4: Area for examination of blood cells and for
A blood smear is examined for: differential cell count in blood smear
• Red cells: Morphology, immature forms, inclusion
bodies, arrangement of cells.
cell distribution, and to select an area for examination of
• White cells: Differential count, abnormal or immature
blood cells. Best morphologic details are seen in the area
forms. where red cells are just touching one another. Low power
• Platelets: Adequacy, abnormal forms.
view is also helpful for the identification of Rouleaux
• Parasites: Malaria, filaria.
formation, autoagglutination of red cells, and micro-
A peripheral blood smear has three parts: Head, body,
filaria. High power objective (45×) is suitable for
and tail (Fig. 22.4). A blood smear should be examined
examination of red cell morphology and for differential
in an orderly manner. Initially, blood smear should be leukocyte count. A rough estimate of total leukocyte
observed under low power objective (10×) to assess count can be obtained which also serves to crosscheck
whether the film is properly spread and stained, to assess the total leukocyte count done by manual counting or
automated method. Oil-immersion objective (100×) is
used for more detailed examination of any abnormal
cells.

Red Cells
Red cells are best examined in an area where they are
just touching one another (towards the tail of the film).
Normal red cells are 7-8 µm in size, round with smooth
contours, and stain deep pink at the periphery and paler
in the center. Area of central pallor is about 1/3rd the
diameter of the red cell. Size of a normal red cell
corresponds roughly with the size of the nucleus of a
small lymphocyte. Normal red cells are described as
normocytic (of normal size) and normochromic (with
normal staining intensity i.e. hemoglobin content).
Morphologic abnormalities of red cells in peripheral
blood smear can be grouped as follows:
Fig. 22.3: Normal blood smear showing red cells (R), a
• Red cells with abnormal size (Fig. 22.5)
neutrophil (N), a lymphocyte (L), eosinophil (E), basophil (B), • Red cells with abnormal staining
monocyte (M), and platelets (P) • Red cells with abnormal shape (Fig. 22.5)
204 Essentials of Clinical Pathology

Fig. 22.5: Variations in size and shape of red cells: (A) Microcytic hypochromic red cells in iron deficiency anemia; (B) Oval
macrocytes and a hypersegmented neutrophil in megaloblastic anemia; (C) Sickle cells in sickle cell anemia; (D) Spherocytes
in hereditary spherocytosis; (E) Fragmented red cells or schistocytes in microangiopathic hemolytic anemia; (F) Target cells
in hemoglobinopathy; (G) Burr cells in chronic renal failure; (H) Tear drop red cells in myelofibrosis; (I) Bite cells and
(J) Blister cell in glucose-6-phosphate dehydrogenase deficiency

• Red cell inclusions (Fig. 22.6)


• Immature red cells (Fig. 22.7)
• Abnormal red cell arrangement (Fig. 22.8).
1. Red cells with abnormal size: Mild variation in red
cell size is normal. Increased variation in red cell size is
called as anisocytosis. This is a feature of most anemias
and is non-specific. Anisocytosis is due to the presence
of microcytes, macrocytes, or both in addition to red cells
of normal size.
Microcytes are red cells smaller in size than normal.
They are seen when hemoglobin synthesis is defective
i.e. in iron deficiency anemia, thalassemias, anemia of
chronic disease, and sideroblastic anemia.
Macrocytes are red cells larger in size than normal. Fig. 22.6: Red cell inclusions: (A) Basophilic stippling;
(B) Howell-Jolly bodies; (C) Pappenheimer bodies; (D) Cabot’s
Oval macrocytes (macro-ovalocytes) are seen in ring
megaloblastic anemia, myelodysplastic syndrome, and
in patients being treated with cancer chemotherapy. hemoglobin content. Red cells with increased area of
Round macrocytes are seen in liver disease, alcoholism, central pallor (i.e. containing less hemoglobin) are called
and hypothyroidism. as hypochromic. They are seen when hemoglobin
2. Red cells with abnormal staining (hemoglobin synthesis is defective, i.e. in iron deficiency, thalassemias,
content): Staining intensity of red cells depends on anaemia of chronic disease, and sideroblastic anemia.
Blood Smear 205

Schistocytes are fragmented red cells, which take


various forms like helmet, crescent, triangle, etc. and
usually have surface projections or spicules. They are
seen in microangiopathic hemolytic anaemia, cardiac
valve prosthesis, and severe burns.
Target cells are red cells with bull's eye appearance.
These red cells show a central stained area and a
Fig. 22.7: Immature red cells: (A) Polychromatic red cell; peripheral stained rim with unstained cytoplasm in
(B) Nucleated red cell
between. They are seen in hemoglobinopathies (e.g.
thalassemias, hemoglobin C disease, sickle cell disease),
obstructive jaundice, and following splenectomy.
Burr cells or echinocytes are small red cells with
regularly placed small projections on surface. They are
seen in uremia.
Acanthocytes are red cells with irregularly spaced
sharp projections of variable length on surface. They are
seen in spur cell anemia of liver disease, McLeod
Fig. 22.8: Abnormal red cell arrangement: (A) Rouleaux phenotype, and following splenectomy.
formation; (B) Autoagglutination
Teardrop cells or dacryocytes have a tapering drop-
like shape. Numerous teardrop red cells are seen in
In dimorphic anemia, there are two distinct popula- myelofibrosis and myelophthisic anemia.
tions of red cells in the same smear. An example is
Blister cells or hemi ghost cells are irregularly
presence of both normochromic and hypochromic red
contracted cells in which hemoglobin is contracted and
cells seen in sideroblastic anemia, iron deficiency anemia
condensed away from the cell membrane. This is seen in
responding to treatment, and following blood transfusion
glucose-6-phosphate dehydrogenase deficiency during
in a patient of hypochromic anemia. In myelodysplastic
acute hemolytic episode.
syndrome, dimorphic picture results from admixture of
Bite cells result from removal of Heinz bodies by the
microcytic hypochromic cells and macrocytes.
pitting action of the spleen (i.e. a part of red cell is bitten
3. Red cells with abnormal shape: Increased variation
off by the splenic macrophages). They are seen in glucose-
in red cell shape is called as poikilocytosis and is a feature
6-phosphate dehydrogenase deficiency and unstable
of many anemias. A red cell that is abnormal in shape is
hemoglobin disease.
called as a poikilocyte.
4. Red cell inclusions: Those inclusions that can be
Sickle cells are narrow and elongated red cells with
one or both ends pointed. Sickle form is assumed when visualized on Romanowsky-stained smears are baso-
a red cell containing hemoglobin S is deprived of oxygen. philic stippling, Howell-Jolly bodies, Pappenheimer
Sickle cells are seen in sickle cell disorders, particularly bodies, and Cabot's rings.
sickle cell anemia. Sickle cells are not seen on blood smear Basophilic stippling or punctate basophilia refers to the
in neonates with sickle cell disease because high presence of numerous, irregular basophilic (purple-blue)
percentage of fetal hemoglobin in red cells prevents granules which are uniformly distributed in the red cell.
sickling. These granules represent aggregates of ribosomes. Their
Spherocytes are red cells, which are slightly smaller in presence is indicative of impaired erythropoiesis and
size than normal, round, stain intensely, and do not have they are seen in thalassemias, megaloblastic anemia,
central area of pallor. The surface area of spherocytes is heavy metal poisoning (e.g. lead), and liver disease.
less as compared to the volume. They are seen in Howell-Jolly bodies are small, round, purple-staining
hereditary spherocytosis, autoimmune hemolytic anemia nuclear remnants located peripherally in red cells. They
(warm antibody type), and ABO hemolytic disease of are seen in megaloblastic anemia, thalassemias,
newborn. hemolytic anemia, and following splenectomy.
206 Essentials of Clinical Pathology

Pappenheimer bodies are basophilic, small, iron- Box 22.2: Role of blood smear in anemias
containing granules in red cells. They give positive Perl's
Prussian blue reaction. Unlike basophilic stippling, • Macrocytic anemia: Differential diagnosis between
Pappenheimer bodies are few in number and are not megaloblastic anemia (oval macrocytosis and hyper-
distributed throughout the red cell. They are seen segmented neutrophils), liver disease (round macrocytosis
following splenectomy and in thalassemias and and target cells), hemolytic anemia (numerous poly-
chromatic cells), and myelodysplastic syndrome (dimor-
sideroblastic anemia.
phic red cells, pseudo-Pelger-Huet neutrophils, giant
Cabot's rings are fine, reddish-purple or red, ring-like platelets, occasional blast cell).
structures. They appear like loops or figure of eight • Microcytic anemias: Differential diagnosis between iron
structures. They indicate impaired erythropoiesis and are deficiency anemia (microcytic hypochromic red cells,
seen in megaloblastic anemia and lead poisoning. pencil cells), thalassemia minor (microcytic hypochromic
5. Immature red cells: red cells, basophilic stippling), and sideroblastic anemia
Polychromatic cells are young red cells containing (dimorphic anemia).
remnants of ribonucleic acid. These cells are slightly • Sickle cell disease: Differentaition between sickle cell trait
larger than normal red cells and have a diffuse bluish- (target cells with no sickle cells), sickle cell anemia (sickle
cells), and sickle cell β-thalassemia (microcytic hypo-
grey tint. (They represent reticulocytes when stained with
chromic red cells, sickle cells).
a supravital stain like new methylene blue). Poly-
• Hemolytic anemias: Spherocytosis (hereditary sphero-
chromasia is due to the uptake of acid stain by cytosis, autoimmune hemolytic anemia), fragmented cells
hemoglobin and basic stain by ribonucleic acid. Presence (microangiopathic haemolytic anemia), bite cells or blister
of polychromatic cells is indicative of active erythro- cells (Glucose-6-phosphate dehydrogenase deficiency),
poiesis and are increased in hemolytic anemia, acute and autoagglutination of red cells (cold hemagglutinin
blood loss, and following specific therapy for nutritional disease).
anemia.
Nucleated red cells are red cell precursors (erythro-
blasts), which are released prematurely in peripheral
blood from the bone marrow. They are a normal finding
in cord blood of newborns. Large number of nucleated
red cells in blood smear is seen in hemolytic disease of
newborn, hemolytic anemia, leukemias, myelophthisic
anemia, and myelofibrosis.
6. Abnormal red cell arrangement:
Rouleaux formation refers to alignment of red cells on top
of each other like a stack of coins. It occurs in multiple
myeloma, Waldenström's macroglobulinemia, hyper-
gammaglobulinemia, and hyperfibrinogenemia.
Autoagglutination refers to the clumping of red cells
in large, irregular groups on blood smear. It is seen in
cold agglutinin disease.
Role of blood smear in anemia is shown in Box 22.2
and Figures 22.9 to 22.11.

White Cells
Approximate idea about total leukocyte count can be
gained from the examination of the smear under high Fig. 22.9: Differential diagnosis of macrocytic anemia on blood
power objective (40× or 45×). A differential leukocyte smear: (A) Megaloblastic anemia; (B) Hemolytic anemia;
count should be carried out. Abnormal appearing white (C) Liver disease; (D) Myelodysplastic syndrome
Blood Smear 207

phil shows non-segmented U-shaped nucleus of


even width. Normally band neutrophils comprise
less than 3% of all leukocytes. Majority of
neutrophils have 3 lobes, while less than 5% have
5 lobes.
In females, 2-3% of neutrophils show a small
projection (called drumstick) on the nuclear lobe.
It represents one inactivated X chromosome.
ii. Eosinophil: Eosinophils are slightly larger than
neutrophils (15-16 µm). The nucleus is often
bilobed and the cytoplasm is packed with
numerous, large, bright orange-red granules. On
blood smears, some of the eosinophils are often
ruptured.
iii. Basophils: Basophils are seen rarely on normal
smears. They are small (9-12 µm), round to oval
cells, which contain very large, coarse, deep purple
granules. It is difficult to make out the nucleus since
granules cover it.
Fig. 22.10: Differential diagnosis of microcytic anemia on iv. Monocytes: Monocyte is the largest of the leuko-
blood smear: (A) Iron deficiency anemia; (B) Thalassemia cytes (15-20 µm). It is irregular in shape, with oval
minor; (C) Thalassemia major; (D) Sideroblastic anemia
or clefted (kidney-shaped) nucleus and fine,
delicate chromatin. Cytoplasm is abundant, blue-
gray with ground glass appearance and often
contains fine azurophil granules and vacuoles.
After migration to the tissues from blood, they are
called as macrophages.
v. Lymphocytes: On peripheral blood smear, two types
of lymphocytes are distinguished: small and large.
The majority of lymphocytes are small (7-8 µm).
Fig. 22.11: Differential diagnosis of hemolytic anemia on
blood smear. (A) Microangiopathic hemolytic anemia showing These cells have a high nuclear-cytoplasmic ratio
fragmented red cells, (B) Hereditary spherocytosis showing with a thin rim of deep blue cytoplasm. The
spherocytes and a polychromatic red cell, and (C) Glucose- nucleus is round or slightly clefted with coarsely
6-phosphate dehydrogenase deficiency showing a blister cell clumped chromatin. Large lymphocytes (10-15
and a bite cell
µm) have a more abundant, pale blue cytoplasm,
which may contain a few azurophil granules.
cells are evaluated under oil-immersion objective. Nucleus is oval or round and often placed on one
1. Morphology of normal leukocytes (refer to Fig. 16.9 side of the cell.
in Chapter 16: Hematopoiesis): 2. Morphology of abnormal leukocytes (Fig. 22.12):
i. Polymorphonuclear neutrophil: Neutrophil measures i. Toxic granules: These are darkly staining, blue-
14-15 µm in size. Its cytoplasm is colorless or lightly purple, coarse granules in the cytoplasm of
eosinophilic and contains multiple, small, fine, neutrophils. They are commonly seen in severe
mauve granules. Nucleus has 2-5 lobes that are bacterial infections.
connected by fine chromatin strands. Nuclear ii. Döhle inclusion bodies: These are small, oval, pale
chromatin is condensed and stains deep purple in blue cytoplasmic inclusions in the periphery of
color. A segmented neutrophil has at least 2 lobes neutrophils. They represent remnants of riboso-
connected by a chromatin strand. A band neutro- mes and rough endoplasmic reticulum. They are
208 Essentials of Clinical Pathology

and metamyelocytes) in peripheral blood and


occurs in infections and inflammatory disorders.
v. Hypersegmented neutrophils: Hypersegmentation of
neutrophils is said to be present when >5% of
neutrophils have 5 or more lobes. They are large
in size and are also called as macropolycytes. They
are seen in folate or vitamin B12 deficiency and
represent one of the earliest signs.
vi. Pelger-Huet cells: In Pelger-Huet anomaly (a benign
autosomal dominant condition), there is failure
of nuclear segmentation of granulocytes so that
nuclei are rod-like, round, or have two segments.
Such granulocytes are also observed in myelo-
proliferative disorders (pseudo-Pelger-Huet cells).
vii. Atypical lymphocytes: These are seen in viral
infections, especially infectious mononucleosis.
Atypical lymphocytes are large, irregularly
shaped lymphocytes with abundant cytoplasm
and irregular nuclei. Cytoplasm shows deep
basophilia at the edges and scalloping of borders.
Nuclear chromatin is less dense and occasional
Fig. 22.12: Morphological abnormalities of white blood cells: nucleolus may be present.
(A) Toxic granules; (B) Döhle inclusion body; (C) Shift to left viii. Blast cells: These are most premature of the
in neutrophil series; (D) Hypersegmented neutrophil in leukocytes. They are large (15-25 µm), round to
megaloblastic anemia; (E) Atypical lymphocyte in infectious oval cells, with high nuclear cytoplasmic ratio.
mononucleosis; (F) Blast cell in acute leukemia Nucleus shows one or more nucleoli and nuclear
chromatin is immature. These cells are seen in
severe infections, infiltrative disorders, and
often associated with toxic granules and are seen leukemia. In leukemia and lymphoma, blood
in bacterial infections. smear suggests the diagnosis or differential
iii. Cytoplasmic vacuoles: Vacuoles in neutrophils are diagnosis and helps in ordering further tests
indicative of phagocytosis and are seen in bacterial (Fig. 22.13 and Box 22.3).
infections. 3. Differential leukocyte count (DLC): DLC refers to
iv. Shift to left of neutrophils: This refers to presence of relative proportion of different leukocytes expressed
immature cells of neutrophil series (band forms as a percentage.

Fig. 22.13: Comparison of blood smears in (A) acute myeloid leukemia and (B) acute lymphoblastic leukemia
Blood Smear 209

Box 22.3: Role of blood smear in leukemia

• It suggests the diagnosis or differential diagnosis


• It suggests further investigations to be performed for
definitive diagnosis.

Principle: Leukocytes are counted on a blood smear and


percentage of each type of leukocyte is recorded.
Uses of DLC
1. To support the diagnosis of infectious, inflammatory,
or allergic disorders.
2. Diagnosis of malignant blood disorders.
Using a high power objective, leukocytes should be
Fig. 22.14: Numerical abnormalities of white blood cells: (A)
classified and counted moving from one field to another
Neutrophilia; (B) Eosinophilia; (C) Monocytosis; (D) Lympho-
as shown in Figure 22.4. DLC should not be carried out cytosis
on the edges and in the tail of the film. In a badly spread
smear, polymorphonuclear neutrophils, monocytes, and 8. Malignant tumors
abnormal cells tend to accumulate in 'feather edge' (tail) 9. Physiologic causes: Exercise, labor, pregnancy,
and lateral edges, while lymphocytes accumulate in the emotional stress.
body of the smear. Also, cells in the body of the smear
are shrunken and darkly stained. DLC on such a smear Leukemoid reaction: This refers to the presence of markedly
will not be representative. If count is high and abnormal increased total leukocyte count (>50,000/cmm) with
cells are present, DLC should be done in the area just immature cells in peripheral blood resembling leukaemia
before the tail of the film. This is because morphology is but occurring in non-leukemic disorders (Fig. 22.15). Its
better appreciated in this area. causes are:
4. Numerical abnormalities of leukocytes (Fig. 22.14): • Severe bacterial infections, e.g. septicemia, pneumo-
For meaningful interpretation, absolute count of nia
leukocytes should be reported. These are obtained as • Severe hemorrhage
follows: • Severe acute hemolysis

Absolute leukocyte count = Percentage of leukocyte ×


Total leukocyte count/ml

Neutrophilia
An absolute neutrophil count greater than 7500/µl is
termed as neutrophilia or neutrophilic leukocytosis.
Causes
1. Acute bacterial infections: Abscess, pneumonia,
meningitis, septicemia, acute rheumatic fever, urinary
tract infection.
2. Tissue necrosis: Burns, injury, myocardial infarction.
3. Acute blood loss
4. Acute hemorrhage
5. Myeloproliferative disorders
6. Metabolic disorders: Uremia, acidosis, gout
7. Poisoning Fig. 22.15: Leukemoid reaction in blood smear
210 Essentials of Clinical Pathology

Table 22.1: Differences between leukemoid reaction and leukemia


Parameter Leukemoid reaction Leukemia

1. Clinical presentation Features of underlying Splenomegaly


disease; fever common
2. Examination of blood
a. Total leukocyte count < 50000/µl Variable, usually > 1 lac/µl
b. Course of neutrophilia Disappears with resolution of Progressive increase
underlying disease
c. Evidence of infection Toxic granules, Döhle Absent
inclusion bodies
d. Basophilia Absent Present
e. Immature cells Few; cells up to myelocyte stage Many; cells up to blasts
3. Examination of marrow Myeloid hyperplasia Increased blasts and immature
cells of neutrophil series;
Suppression of other cell lines
4. Clonality Polyclonal Monoclonal
5. Karyotype Normal Abnormal

• Poisoning II. Increased destruction in peripheral blood:


• Burns 1. Neonatal isoimmune neutropaenia
• Carcinoma metastatic to bone marrow 2. Systemic lupus erythematosus
Leukemoid reaction should be differentiated from 3. Felty's syndrome
chronic myeloid leukemia (Table 22.1). III. Increased sequestration in spleen:
1. Hypersplenism.
Neutropenia: Absolute neutrophil count less than 2000/
µl is neutropenia. It is graded as mild (2000-1000/µl), Eosinophilia
moderate (1000-500/µl), and severe (< 500/µl). This refers to absolute eosinophil count greater than 600/
µl.
Causes Causes
I. Decreased or ineffective production in bone marrow: 1. Allergic diseases: Bronchial asthma, rhinitis, urticaria,
1. Infections drugs.
a. Bacterial: typhoid, paratyphoid, miliary tuber- 2. Skin diseases: Eczema, pemphigus, dermatitis
culosis, septicemia herpetiformis.
b. Viral: influenza, measles, rubella, infectious 3. Parasitic infection with tissue invasion: Filariasis,
mononucleosis, infective hepatitis. trichinosis, echinococcosis.
c. Protozoal: malaria, kala azar 4. Hematologic disorders: Chronic myeloproliferative
d. Overwhelming infection by any organism disorders, Hodgkin's disease, peripheral T cell
2. Hematologic disorders: megaloblastic anemia, lymphoma.
aplastic anemia, aleukemic leukemia, myelo- 5. Carcinoma with necrosis.
phthisis. 6. Radiation therapy.
3. Drugs: 7. Lung diseases: Loeffler's syndrome, tropical eosino-
a. Idiosyncratic action: Analgesics, antibiotics, philia
sulfonamides, phenothiazines, antithyroid 8. Hypereosinophilic syndrome.
drugs, anticonvulsants. Basophilia: Increased numbers of basophils in blood
b. Dose-related: Anticancer drugs (>100/μl) occurs in chronic myeloid leukemia, poly-
4. Ionizing radiation cythemia vera, idiopathic myelofibrosis, basophilic
5. Congenital disorders: Kostman's syndrome, cyclic leukemia, myxedema, and hypersensitivity to food or
neutropenia, reticular dysgenesis. drugs.
Blood Smear 211

Monocytosis
Box 22.4: Differential diagnosis of lymphocytosis
This is an increase in the absolute monocyte count above
1000/µl. • Mature lymphocytosis: Viral infections, whooping
cough, tuberculosis, infectious lymphocytosis, chronic
Causes lymphocytic leukemia
1. Infections: Tuberculosis, subacute bacterial endocar- • Atypical lymphocytosis: Infectious mononucleosis,
ditis, malaria, kala azar. cytomegalovirus, toxoplasmosis, infectious hepatitis
2. Recovery from neutropenia. • Lymphoblasts: Acute lymphoblastic leukemia
3. Autoimmune disorders.
4. Hematologic diseases: Myeloproliferative disorders,
fingerstick), they occur in clumps. If platelet count is done
monocytic leukemia, Hodgkin's disease.
on automated blood cell counters using EDTA-anticoagu-
5. Others: Chronic ulcerative colitis, Crohn's disease,
lated blood sample, about 1% of persons show falsely
sarcoidosis.
low count due to the presence in them of EDTA-
Lymphocytosis dependent antiplatelet antibody.
Examination of a parallel blood film is useful in
This is an increase in absolute lymphocyte count above
avoiding the false diagnosis of thrombocytopenia in such
upper limit of normal for age (4000/µl in adults, >7200/
µl in adolescents, >9000/µl in children and infants) (Box cases. Occasionally, platelets show rosetting around
neutrophils (platelet satellitism) (Fig. 22.16). This is seen
22.4).
in patients with platelet antibodies and in apparently
Causes normal persons.
1. Infections: Blood smear examination can be helpful in deter-
• Viral: Acute infectious lymphocytosis, infective mining underlying cause of thrombocytopenia such as
hepatitis, cytomegalovirus, mumps, rubella, leukemia, lymphoma, or microangiopathic hemolytic
varicella anemia (Box 22.5).
• Bacterial: Pertussis, tuberculosis
• Protozoal: Toxoplasmosis Organisms
2. Hematological disorders: Acute lymphoblastic Common parasites seen in blood are malaria parasites
leukemia, chronic lymphocytic leukemia, multiple and microfilaria (See Chapter 26: Diagnosis of Malaria
myeloma, lymphoma. and Other Parasites in Blood). Other parasites that can
3. Other: Serum sickness, post-vaccination, drug
reactions. Box 22.5: Role of blood smear in thrombocytopenia

Platelets It suggests probable cause, e.g. leukemia, microangio-


Platelets are small, 1-3 µm in diameter, purple structures pathic process like thrombotic thrombocytopenic purpura
or disseminated intravascular coagulation, aplastic
with tiny irregular projections on surface. In blood films
anemia, May-Hegglin anomaly.
prepared from non-anticoagulated blood (i.e. direct

Fig. 22.16: Causes of false thrombocytopenia on automated hematology analyzer:


(A) Clumps of platelets; (B) Platelet satellitism
212 Essentials of Clinical Pathology

be seen in blood smear are Leishmania donovani, and • Eosinophils: 1-6%


trypanosomes. Sometimes bacteria such as meningococci • Basophils: 0-1%
and fungi (e.g. Histoplasma capsulatum) can be observed
within phagocytic leukocytes. Critical Values
• Blood smear showing sickle cells
REFERENCE RANGES • Blood smear showing blast cells
• Leukemoid reaction
Normal Blood Smear • Suspected aplastic anemia
• Malarial parasites
Red blood cells: Normocytic and normochromic
• Absolute neutrophil count <500/cmm.
White blood cells: Total and differential leucocyte
counts within normal limits BIBLIOGRAPHY
Platelets: Adequate 1. Bain BJ. Diagnosis from the blood smear. N Engl J Med
Parasites: Not seen. 2005;353:498-507.
2. Houwen B. Blood film preparation and staining
Differential Leukocyte Count procedures. Clin Lab Med 2002;22:1-14.
3. International Council for Standardization in Haema-
• Neutrophils: 40-75% tology (ICSH). ICSH reference method for staining of
• Lymphocytes: 20-40% (in children between 4 months blood and bone marrow by azure B and eosin Y
to 4 years of age, percentage of lymphocytes is more (Romanowsky stain). Br J Haematol 1984;57:707-10.
4. Kawthalkar SM. Essentials of Hematology. New Delhi.
than neutrophils; this is called as inverted differential Jaypee Brothers Medical Publishers (P) Ltd, 2006.
in children) 5. Woronzoff-Dashkoff KK. The Wright-Giemsa stain:
• Monocytes: 2-10% secrets revealed. Clin Lab Med 2002;22:15-23.
23

Red Cell Indices

Red cell indices are mean cell volume (MCV), mean cell MCV is expressed in femtoliters or fl (10–15 of a liter).
hemoglobin (MCH), and mean cell hemoglobin concen- It corresponds with red cell diameter on blood smear.
tration (MCHC). They are also called as “absolute Normal MCV is 80-100 fl.
values”. They are derived from the values of hemoglobin,
packed cell volume (PCV or hematocrit), and red cell Causes of Increased MCV
count. Red cell indices are accurately measured by • Megaloblastic anemia
automated hematology analyzers. Recently, a new • Non-megaloblastic macrocytosis: Chronic alcoholism,
parameter called red cell distribution width (RDW) has liver disease, hypothyroidism, normal pregnancy,
been introduced. reticulocytosis
• Newborns.
USES OF RED CELL INDICES
Causes of Low MCV
1. Morphological classification of anemia: Based on
values of red cell indices, anemia is classified into • Microcytic hypochromic anemia
threee main types: normocytic normochromic, MCV is normal in normocytic normochromic anemia
microcytic hypochromic, and macrocytic normo- (acute blood loss, hemolysis, aplastic anemia).
chromic (See Chapter 27: Laboratory Tests in In the presence of large number of abnormal red cells
Anemia). Calculation of red cell indices is especially like sickle cells, and in dimorphic anemia (e.g. mixed
helpful in mild or moderate anemia when red cell normocytic and microcytic), MCV may be normal (since
changes are subtle and often difficult to appreciate it is an average value) and thus unreliable for morpho-
on stained blood smear. logical classification.
2. Differentiation of iron deficiency anemia from Mentzer index is derived by dividing MCV with red
thalassemia trait: In iron deficiency, MCV, MCH, and cell count. Ratio of less than 13 is seen in thalassemia
MCHC are low, while in thalassemia trait, MCV and while ratio is more than 13 in iron deficiency anemia.
MCH are low and MCHC is normal.
MEAN CELL HEMOGLOBIN (MCH)
MEAN CELL VOLUME
MCH is the average amount of hemoglobin in a single
MCV is a measure of average size of the red cells. It is
red cell. It is obtained by dividing hemoglobin value by
measured directly by automated instruments from the
red cell count.
measurement of volume of each red cell. With semi-
automated instruments and by manual method, it is
obtained by dividing PCV by red cell count. Hemoglobin in grams/dl
MCH = ——————————————— × 10
Red cell count in millions/cmm
PCV in% MCH is expressed in picograms or pg (10–12 gram).
MCV = —————————————— × 10
Red cell count in million/cmm Reference range is 27-32 pg.
214 Essentials of Clinical Pathology

MCH is decreased in microcytic hypochromic Red Cell Distribution Width (RDW)


anemia, and increased in macrocytic anemia and in Some automated analyzers measure red cell distribution
newborns. width or RDW. It is a measure of degree of variation in
MEAN CELL HEMOGLOBIN red cell size (anisocytosis) in a blood sample. It is helpful
CONCENTRATION (MCHC) in differential diagnosis of some anemias. Amongst
microcytic anemias, RDW is low in β thalassemia trait,
MCHC is obtained by dividing hemoglobin value by PCV
high in iron deficiency anemia, and normal in anemia of
and expressed in grams/dl or grams/liter. It refers to
chronic disease. Normal RDW is 9.0 to 14.5.
concentration of hemoglobin in 1 dl or 1 liter of packed
red cells. REFERENCE RANGES
• Mean cell volume: 80-100 fl
Hemoglobin in grams/dl • Mean cell hemoglobin: 27-32 pg
MCHC = ——————————— × 100
PCV in %
• Mean cell hemoglobin concentration: 30-35 g/dl
• Red cell distribution width: 9.0-14.5

Reference range is 30-35 grams/dl. BIBLIOGRAPHY


MCHC is raised in hereditary spherocytosis, and is
1. Henry JB. Clinical diagnosis and management by
decreased in hypochromic anemia. MCHC corresponds laboratory methods (20th Ed). Philadelphia: WB
with degree of hemoglobinization of red cells on a blood Saunders Company, 2001.
smear. If MCHC is normal, red cell is normochromic, 2. Wallach J. Interpretation of Diagnostic Tests (7th Ed).
and if low, red cell is hypochromic. Philadelphia: Lippincott Williams and Wilkins, 2000.

tahir99 - UnitedVRG
vip.persianss.ir
24
Erythrocyte
Sedimentation Rate

The erythrocyte sedimentation rate (ESR) measures the


Box 24.1: Factors affecting erythrocyte
rate of settling (sedimentation) of erythrocytes in
sedimentation rate
anticoagulated whole blood. Anticoagulated blood is
allowed to stand in a glass tube for 1 hour and the length • Factors increasing ESR
of column of plasma above the red cells is measured in – Old age
millimeters; this corresponds to ESR. – Pregnancy
– Anemia
– Elevated fibrinogen
STAGES OF ERYTHROCYTE
– Macrocytosis
SEDIMENTATION RATE – Technical factors: High temperature, tilting of ESR tube
There are three stages of erythrocyte sedimentation: • Factors decreasing ESR
• Stage 1: Formation of rouleaux or lag phase (10 – Microcytosis
minutes): Red cells stack together like a pack of coins. – Low fibrinogen
The ESR depends mainly on this stage. – Polycythemia
– Marked leukocytosis
• Stage 2: Sinking of rouleaux or decantation phase (40 – Technical factors: Vibration of tube during test
minutes): Rapid and constant sedimentation. The
longer the tube, the longer is this stage and higher
the value of ESR. other acute phase proteins (C-reactive protein,
• Stage 3: Packing of rouleaux (10 minutes): Slow haptoglobin, ceruloplasmin, α1-antitrypsin, etc.) and
sedimentation. immunoglobulins increase ESR. Increased proteins
in plasma reduce negative charge on the surface of
FACTORS AFFECTING ERYTHROCYTE red cells and reduce the zeta potential (the electrical
SEDIMENTATION RATE repulsion between red cells); this brings red cells
closer together and facilitates rouleaux formation.
Red cells, composition of plasma, and technical factors
Removal of fibrinogen by defibrination and increase
affect ESR (Box 24.1).
of albumin retard ESR.
1. Red cells: Alteration of ratio of red cells to plasma
3. Technical factors: ESR increases with room tempe-
affects ESR. Decreased red cell mass in anemia
rature. Tilting of the tube, and length and bore of the
increases ESR. Conversely, increased red cell mass
tube affect ESR.
in polycythemia decreases ESR.
Macrocytes tend to sediment rapidly than microcytes.
SIGNIFICANCE OF ERYTHROCYTE
Sickle cells and spherocytes are unable to form
SEDIMENTATION RATE
rouleaux and therefore ESR is low in sickle cell disease
and hereditary spherocytosis. In these conditions, ESR is elevated in a wide range of organic diseases. ESR
ESR is not reliable as an indicator of illness. is not a specific and diagnostic test for any disease.
2. Plasma: The most important factor affecting ESR is However, it is helpful in differentiating functional from
the composition of plasma. Increase in fibrinogen, organic disease. Raised ESR signifies presence of some

tahir99 - UnitedVRG
vip.persianss.ir
216 Essentials of Clinical Pathology

Table 24.1: Causes of increased erythrocyte (≥100 mm at 1 hour) is seen in infections, parapro-
sedimentation rate teinemias, metastatic malignancy, polymyalgia rheu-
1. Infections • Acute rheumatic fever matica, temporal arteritis, and rheumatoid arthritis.
• Osteomyelitis ESR is decreased in polycythemia, congestive cardiac
• Bacterial endocarditis failure, dehydration, sickle cell anemia, hereditary
• Pyogenic arthritis spherocytosis, and hypofibrinogenemia.
• Pelvic inflammatory disease
In patients with chronic inflammatory conditions like
• Tuberculosis
• Acute hepatitis rheumatoid arthritis, ESR can be helpful in distinguishing
iron deficiency anemia from anemia of chronic disease.
2. Inflammatory diseases • Rheumatoid arthritis
In inflammatory conditions, ESR rises more than 24
• Systemic lupus
erythematosus hours after onset and returns to normal about 4 weeks
• Temporal arteritis following resolution.
• Polymyalgia rheumatica There is no role of ESR in the evaluation of asympto-
3. Acute myocardial matic patients.
infarction
4. Malignancy
INDICATIONS FOR MEASUREMENT OF
ERYTHROCYTE SEDIMENTATION RATE
5. Paraproteinemias • Multiple myeloma
• Waldenström’s It is recommended to measure ESR (i) when infectious,
macroglobulinemia inflammatory, or a neoplastic disease is suspected in
• Cryoglobulinemia symptomatic individuals in whom a specific diagnosis
6. Technical problems • Increased temperature has not been established, (ii) to monitor disease activity
• Tilted ESR tube in tuberculosis, inflammatory arthritis, rheumatic fever,
7. Others • Ruptured ectopic pregnancy Hodgkin’s disease, giant cell arteritis, and polymyalgia
• Anemia rheumatica, and (iii) as a diagnostic criterion for temporal
• Renal disease with arteritis and polymyalgia rheumatica. ESR can also be
azotemia helpful in distinguishing iron deficiency anemia from
• Administration of dextran or anemia of chronic disease in patients known to have
oral contraceptives inflammatory disease.

METHODS FOR ESTIMATION OF


organic disease, which needs evaluation. Most of the ERYTHROCYTE SEDIMENTATION RATE
inflammatory and neoplastic diseases are associated with
an increase in ESR (Table 24.1). These include:
ESR correlates with disease activity and therefore it • Westergren method
is helpful in monitoring disease activity and response to • Wintrobe method
therapy in acute rheumatic fever, bacterial endocarditis, • Zeta sedimentation ratio
tuberculosis, rheumatoid arthritis, temporal arteritis, • Micro-ESR
polymyalgia rheumatica, and Hodgkin’s disease.
ESR is an important criterion in establishing the Westergren Method
diagnosis of temporal arteritis and polymyalgia International Council for Standardization in Hematology
rheumatica.
recommends Westergren method for estimation of ESR.
ESR is not significantly raised in typhoid fever,
malaria, infectious mononucleosis, angina pectoris, Equipment and Reagent
osteoarthritis, acute appendicitis, peptic ulcer, acute
allergy, and unruptured ectopic pregnancy. In emer- 1. Westergren ESR tube: This is a straight glass pipette
gency situations, ESR may be helpful in distinguishing measuring 300 mm in length and calibrated in mm
angina pectoris from myocardial infarction, and acute from 0-200 (top to bottom). The markings are only
appendicitis from other causes of acute abdomen over the lower 2/3rds of the tube. The tube is open-
particularly pelvic inflammatory disease or ruptured ended. Internal diameter should not be less than 2.55
ectopic pregnancy. Very high or extreme elevation of ESR mm (Fig. 24.1). The tube should be clean and dry.

tahir99 - UnitedVRG
vip.persianss.ir
Erythrocyte Sedimentation Rate 217

Fig. 24.1: Westergren and Wintrobe tubes for measurement of ESR

Specimen: Venous blood is collected in trisodium citrate


solution in 4:1 (blood: citrate) proportion. If specimen is
kept at room temperature, test should be carried out
within 4 hours of blood collection; if stored at 4°C, a delay
of up to 6 hours is permissible. Blood anticoagulated with
EDTA can be tested within 24 hours if stored at 4°C
(provided it is diluted with trisodium citrate before
testing).

Method
1. Mix anticoagulated blood sample thoroughly. The
Westergren tube is filled with the blood sample up
to the “0” mark. A rubber bulb or a mechanical device
should be used for filling. There should be no air
bubbles in the blood.
2. The tube is placed in a strictly vertical position in the
Fig. 24.2: Westergren ESR stand with ESR tubes ESR stand and left undisturbed for 1 hour. It should
not be kept in direct sunlight and should not be
subjected to vibrations.
2. Westergren stand: This holds the tube in a motionless, 3. After exactly 1 hour, read the height of the column of
vertical position (Fig. 24.2). plasma above the red cell column in mm.
3. Anticoagulant-diluent solution: Trisodium citrate 4. Express the result as:
dihydrate is the anticoagulant of choice. Its compo-
sition is as follows:
Erythrocyte sedimentation rate = ——— mm in 1 hour.
Trisodium citrate dihydrate 32.08 gm
Distilled water upto 1000 ml Precautions
It is filtered through a sterile membrane (0.22 μm)
into a sterile container and stored in a refrigerator at 4°C. 1. Use the correct proportion of blood and anticoagulant.
It keeps for several months, but if it becomes turbid (due Mix blood and anticoagulant thoroughly. There
to the growth of moulds), it should be discarded. should be no clots and air bubbles in blood.
Alternate anticoagulant is EDTA (1.5 mg/ml); 2. The reference range relates to test performed at room
however, such anticoagulated blood sample should be temperature of 18-25°C. If temperature is higher, ESR
diluted with trisodium citrate just before the test will increase and different reference range will have
(1 volume of trisodium citrate to 4 volumes of blood). to be derived.

tahir99 - UnitedVRG
vip.persianss.ir
218 Essentials of Clinical Pathology

3. ESR tube must be kept vertically. Even a slight tilting


will increase the ESR.

Other Methods

Wintrobe Method
Wintrobe tube (Fig. 24.1) can be used for estimation of
both ESR and packed cell volume (PCV). After obtaining
ESR in the first hour, the tube can be spun in a centrifuge
to get PCV. Wintrobe’s method is more reliable when
ESR is low, while Westergren’s method is more sensitive
for high ESR. EDTA or double oxalate is used as an
anticoagulant. Length of Wintrobe tube is shorter (110
mm) and internal diameter is about 3 mm.

Zeta Sedimentation Ratio (ZSR)


A special device called as zetafuge is required. ZSR is
not affected by anemia, unlike Westergren method.

Micro-ESR Fig. 24.3: Formation of acute phase proteins by


liver in inflammation
This method uses capillary blood that is collected in
anticoagulant-coated capillary tubes. It is more suitable
protein. Test for CRP is helpful in determining the
in small children. presence and extent of inflammation and in monitoring
response to therapy. Among the acute phase reactants,
ACUTE PHASE REACTANTS CRP is earliest to rise during inflammation and to return
The acute phase proteins are plasma proteins synthesized rapidly to normal level following successful therapy.
in liver that undergo change in concentration in response Unlike other acute phase proteins, hormones or drugs
to infection, tissue injury, and inflammation (Table 24.2). do not affect CRP levels and no deficiency state of CRP
Acute phase proteins are synthesized by liver has been reported. Also, CRP rises to a more significantly
following stimulation by cytokines like interleukin-1, higher level than other acute phase reactants. Therefore
interleukin-6, and tumor necrosis factor-α synthesized among acute phase proteins, measurement of CRP is
by monocytes and macrophages (Fig. 24.3). Acute phase preferred.
reactants play a role in the defensive process of In acute disease, CRP level correlates well with ESR.
inflammation. They help in opsonization, neutralization As compared to ESR, CRP is (i) earlier to rise in
of proteolytic enzymes released from neutrophils inflammation and earlier to return to normal following
(thereby limiting tissue damage), clearance of cell debris, resolution, (ii) more sensitive, and (iii) more specific since
and healing of wounds it is not affected by anemia, polycythemia, alteration of
Tests are available for estimation of C-reactive protein red cell shape, and paraproteinemia (Table 24.3).
(CRP) in serum. When it was first described in 1930, it Normal level of CRP is less than 8 mg/L.
was found to bind to C-polysaccharide in the cell wall of
Uses of C-reactive Protein
Streptococcus pneumonie; hence the name C-reactive
1. Detection of infection: CRP level is elevated in bacterial
infections. It is especially helpful in neonates (bacterial
Table 24.2: Acute phase proteins
sepsis and meningitis). Sequential testing is helpful
• C-reactive protein • Serum amyloid A protein in monitoring response to antimicrobial therapy. In
• Fibrinogen • Ceruloplasmin immunocompromised patients, due to absence of
• α1-antitrypsin • α1 acid glycoprotein
fever and leukocytosis, elevation of CRP is helpful in
• Haptoglobin • Ferritin
suggesting infection.

tahir99 - UnitedVRG
vip.persianss.ir
Erythrocyte Sedimentation Rate 219

Table 24.3: Comparison of erythrocyte sedimentation rate and C-reactive protein

Erythrocyte sedimentation rate C-reactive protein

1. Affected by age, sex, pregnancy, temperature, 1. Not affected


level of plasma proteins, anemia and red cell morphology
2. Less sensitive and specific for acute phase response 2. More sensitive and more specific for acute phase
response
3. Inexpensive and easy procedure 3. Expensive and requires sophisticated materials and
equipment
4. Specimen: Fresh whole anticoagulated blood 4. Specimen: Serum that may have been stored
5. Indirect means of assessing acute phase response 5. Direct measurement of acute phase response

2. Assessment of severity of inflammatory disorders like Erythrocyte Sedimentation Rate by


rheumatoid arthritis and acute rheumatic fever. Wintrobe Method
3. Detection of postoperative infection: CRP level begins to • Males: 0-9 mm in 1 hour
rise 4-6 hours after surgery, peaks in 2-3 days, and • Females: 0-20 mm in 1 hour
returns to normal by 7th day. Failure of expected • Children: 0-13 mm in 1 hour
normalization indicates postoperative infection.
4. Myocardial infarction: CRP rises over 24-48 hours, C-reactive Protein
peaks at 72 hours, and disappears by 7th day after < 8 mg/L.
acute myocardial infarction. Failure to return to
normal indicates re-infarction. BIBLIOGRAPHY
5. Screening for presence or absence of organic disease. 1. Brigden ML. Clinical utility of the erythrocyte
sedimentation rate. Am Fam Physician 1999;60:
REFERENCE RANGES 1443-50.
2. International Council for Standardization in Hema-
Erythrocyte Sedimentation Rate by tology. ICSH recommendation for measurement of
erythrocyte sedimentation rate. J Clin Path 1993;46:
Westergren Method
198-203.
• Males < 50 years: 0-15 mm in 1 hr 3. Smellie WS, Forth JO, McNulty CAM, et al. Best practice
• Females<50 years: 0-20 mm in 1 hr in primary care pathology: review 2. J Clin Pathol
2006;59:113-20.
• Children: 0-10 mm in 1 hr 4. Stuart J and Lewis SM. Recommendations, safety and
• Elderly (>50 years): Males: 0-20 mm in 1 hr and quality control of erythrocyte sedimentation rate. World
females: 0-30 mm in 1 hr. Health Organization. 1993. WHO/LBS/93.1.

tahir99 - UnitedVRG
vip.persianss.ir
25
Examination of
Bone Marrow

NORMAL BONE MARROW development, mature blood cells leave the bone marrow
and enter the circulation by passing through or in
Bone marrow is the site of hematopoiesis in postnatal
between the endothelial cells of the sinusoids.
life. Bone marrow is one of the largest organs in the body,
Marrow fibroblasts (reticular cells) are highly
and although distributed widely in the skeleton,
branched cells, which form reticulin fibers and form a
functions as a single unit. It is located within the cavities
supporting framework for the hematopoietic cells.
of the bones and mainly consists of hematopoietic cells,
Accumulation of lipid in reticular cells is said to produce
vascular sinusoids, fibroblasts, fat cells, and macro-
fat cells of the bone marrow.
phages. There are no lymphatic channels in the bone
Amount of fatty tissue depends on activity of
marrow. There are two types of marrow: red and yellow.
hematopoietic cells. The proportion of fat cells increases
Red marrow refers to the active hematopoietic tissue
with advancing age with corresponding decrease in
while fatty tissue comprises the yellow (inactive)
hematopoietic tissue. On demand, fatty tissue (yellow
marrow. Red coloration is due to presence of large
marrow) can be rapidly replaced by hematopoietic tissue
amounts of developing red cells. The average volume of
(red marrow). In children and adolescents, fat cells
bone marrow (red and yellow) in an adult is 3000-4000
occupy about 20-30% of the bone marrow volume, in
ml. Red or active marrow constitutes 1500 ml.
The hematopoietic cells are present as cords between adults about 50%, while in the elderly about 70%.
vascular sinusoids and are supported by a framework of Macrophages in bone marrow serve various functions
branching processes of fibroblasts and reticulin fibers. like synthesis of hematopoietic growth factors and
Erythroid precursors are present as clusters (islands) and inhibitory substances, phagocytosis of senescent red cells,
are closely associated with centrally placed sinusoids. and transport of iron to the erythroblasts.
An erythroid island consists of a centrally placed The stromal cells of the bone marrow (fibroblasts, fat
macrophage around which erythroid precursors are cells, macrophages, and endothelial cells) and vascular
concentrically arranged. Early granulocyte precursors are sinusoids constitute the bone marrow microenvironment,
located close to the bony trabecule, while more mature which is essential for normal hematopoiesis.
granulocytes are located more centrally, between During infancy and early childhood, hematopoiesis
adjacent trabeculae. Megakaryocytes, the largest of the is occurring in almost all the bones of the body. By late
hematopoietic cells, are closely apposed to the walls of childhood, hematopoiesis becomes mainly restricted to
the sinusoids. Lymphocytes and plasma cells are mostly flat bones like sternum, ribs, iliac bones, and vertebre,
present around capillaries and arterial vessels. In contrast and proximal ends of long bones; other sites of red
to other blood cells, lymphocytes also divide and mature marrow are transformed into yellow marrow. However,
outside the bone marrow. when there is increased demand for blood cells, blood
Hematopoiesis occurs extravascularly in between cell production can resume in the yellow marrow. In
interconnecting marrow sinusoids. The marrow sinu- extremely severe cases (e.g. chronic hemolytic anemia),
soids are lined by endothelial cells and are covered resumption of hematopoiesis in other organs like liver
partially by cytoplasmic processes of fibroblasts. After and spleen (extramedullary hematopoiesis) can occur.

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Bone Marrow 221

Apart from its major function of hematopoiesis, bone 2. Suspected acute leukemia: In majority of cases, acute
marrow also plays a role in the removal of senile and leukemia can be diagnosed from examination of a
defective red cells (via macrophages) and immunity blood smear and cell counts. Bone marrow exami-
(processing of antigen by macrophages and synthesis of nation is carried out in acute leukemia for:
antibodies by plasma cells). • Detection of subleukemic or aleukemic leukemia
There are two techniques for sampling of bone • Comparison of baseline marrow smear with
marrow: aspiration and biopsy. In aspiration, bone follow-up marrow aspiration smears during
marrow fluid is obtained by a special needle and syringe. treatment
Smears from this material are prepared on glass slides, • Diagnosis of acute myeloid leukemia with
stained, and examined under the microscope. In bone trilineage dysplasia
marrow trephine biopsy (core biopsy), a small tissue • Cytogenetic analysis.
piece of bone marrow is removed with a special needle, • Detection of remission
processed to obtain histological sections, and examined. 3. Suspected myelodysplastic syndrome.
4. Suspected myeloproliferative disorders including
INDICATIONS FOR BONE MARROW chronic myeloid leukemia, polycythemia vera,
essential thrombocythemia, and myelofibrosis.
EXAMINATION
5. Suspected plasma cell dyscrasia: Bone marrow
Before performing bone marrow aspiration/biopsy, one aspiration is indicated if multiple myeloma is
should assess clinical features, treatment received, and suspected from clinical and radiologic features.
relevant laboratory test results (especially basic blood 6. Suspected chronic lymphoid leukemias like chronic
studies and peripheral blood smear). Based on above lymphocytic leukemia, prolymphocytic leukemia,
data, if appropriate indication exists, then examination hairy cell leukemia, etc.
of bone marrow is carried out and findings are correlated 7. Investigation of pyrexia of unknown origin:
to arrive at the final diagnosis. Sometimes, bone marrow aspiration smears are
helpful in detecting Histoplasma capsulatum and
INDICATIONS FOR BONE MARROW Leishmania donovani organisms in macrophages of
ASPIRATION bone marrow. Aspirated material can also be cultured
1. Unexplained cytopenia: If the cause of cytopenia to isolate above organisms or mycobacteria.
8. Suspected storage disorder like Gaucher’s disease or
(anemia, leukopenia, or thrombocytopenia) is not
Neimann-Pick disease.
apparent from blood investigations and clinical
9. Suspected infections like kala azar, miliary tuber-
details, bone marrow examination is indicated.
culosis, or histoplasmosis.
(A) To distinguish amongst causes of microcytic hypo-
chromic anemia (e.g. iron deficiency from chronic
INDICATIONS FOR BONE MARROW BIOPSY
disease), bone marrow examination can be done to
assess storage iron. Identification of ringed sidero- 1. Repeated failure of aspiration (dry tap) which may
blasts is helpful in diagnosis of sideroblastic anemia be due to faulty technique, myelofibrosis or leuke-
or myelodysplastic syndrome. In macrocytic anemia, mia.
in the absence of vitamin assays and typical features 2. Suspected aplastic anemia
of megaloblastic anemia on blood smear, marrow 3. Suspected myelofibrosis.
aspiration is carried out to distinguish megaloblastic 4. Suspected focal lesions like granuloma, metastatic
macrocytosis from non-megaloblastic one. (B) In deposit, or infiltrate of lymphoma.
leukopenia or thrombocytopenia, bone marrow 5. Suspected hairy cell leukemia.
examination is helpful in distinguishing peripheral 6. Suspected bone disorder, e.g. osteopetrosis.
destruction from deficient production. (C) In the 7. Staging of lymphoma.
presence of isolated thrombocytopenia, if idiopathic
thrombocytopenic purpura (ITP) is suspected, CONTRAINDICATIONS
marrow examination is done for ruling out under-
lying hematologic disorder or deficient platelet Bone marrow aspiration or biopsy is contraindicated in
production. hemophilia and other coagulation disorders; however,

tahir99 - UnitedVRG
vip.persianss.ir
222 Essentials of Clinical Pathology

in these patients it can be performed under cover of


appropriate replacement therapy. Thrombocytopenic
purpura is not a contraindication since applying firm
pressure for 5 minutes at the puncture site can prevent
excess bleeding.

SITES FOR BONE MARROW


ASPIRATION OR BIOPSY
• Iliac spines or crest: The most frequently used site
for both marrow aspiration and biopsy in children
(>1 year of age) as well as in adults is posterior iliac
crest (posterior superior iliac spine). This site has a
large reservoir of marrow, is located just beneath the
skin and therefore easily accessible. Also, there are
no large blood vessels or nerves close to this area,
and as the patient’s back is towards the physician,
patient’s apprehension is less.
In obese patients, anterior superior iliac spine,
Fig. 25.1: Sites of bone marrow aspiration
being more easily localized, can be used.
(shaded areas)
• Sternum: Previously, sternum was commonly used
for aspiration of bone marrow in adults (at the level
of second intercostal space in midline). However, it
is associated with the risk of perforation of posterior
sternal plate and puncturing of underlying large
blood vessels and right atrium with serious conse-
quences. It also causes greatest patient anxiety. This
site should not be used in children as the bone is thin
and marrow cavity is small.
• Spinous processes of lumbar vertebra: This is an
additional site for aspiration in adults.
• Tibia: In infants under 1 year of age, marrow can be
Fig. 25.2: Klima and Salah bone marrow aspiration needles.
aspirated from the medial aspect of upper end of tibia Adjustable guard prevents damage to deeper structures. Stylet
just beneath tibial tuberosity. In older children, the keeps needle patent during introduction
tibial cortical bone becomes hard and cellularity of
marrow decreases, and therefore this site is not used.
solution, clean and dry glass slides, spreader slide,
Sites for bone marrow aspiration are shown in Figure
gloves, drapes, gauze, and a skin antiseptic solution.
25.1.
All aseptic precautions should be observed during
the procedure. Various bone marrow aspiration
METHOD
needles are available. Salah and Klima needles are
Bone Marrow Aspiration commonly used (Fig. 25.2). Salah needle has a guard
with a side screw, while Klima needle has a guard
1. An informed consent should be obtained before the which screws along the length of the needle. Guards
procedure. Bone marrow aspiration or biopsy should on these needles are adjustable to control the depth
be performed by the physician. An assistant is of penetration. The guard may slip from the Salah
required for preparation of smears. needle during the procedure. For aspiration from iliac
2. A sterile tray should be prepared containing crest and tibia, if required, guard may be removed to
autoclaved bone marrow aspiration needle, sterile increase the length of the needle. Guard is essential
disposable syringes with needles, local anesthetic during sternal aspiration to prevent penetration

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Bone Marrow 223

Fig. 25.4: (A) Bone marrow smear and (B) Bone marrow
biopsy sample

6. A 5 or 10 ml syringe is attached to the needle and a


small amount of marrow is aspirated (till the first drop
of blood appears i.e. 0.25-0.50 ml) by quickly pulling
Fig. 25.3: Jamshidi bone marrow biopsy needle the plunger of the syringe. Aspiration is associated
with sharp pain (suction pain). Aspiration of larger
deeper than necessary and avoid damage to heart or amount of blood causes dilution of marrow sample
large vessels. Jamshidi needle, which is longer, can by peripheral blood with subsequent difficulties in
be used for both aspiration and biopsy from iliac crest interpretation of smears. If no material is aspirated,
(Fig. 25.3). stylet is replaced, needle is redirected, and aspiration
3. For aspiration from posterior superior iliac spine, attempted again.
patient should lie on one side with back towards the 7. The syringe should be handed over to the assistant
physician, knees and hips flexed, and the knees for preparation of smears on glass slides. The smears
drawn towards the chest. The site for aspiration should be made promptly, before clotting occurs, by
should be selected and scrubbed with soap and water. putting one drop of the aspirated material near one
After wearing sterile gloves, antiseptic solution is end of a glass slide and spreading it similar to a blood
applied in a circular fashion, moving from center film (Fig. 25.4). Before making smears, any excess
towards periphery. A sterile drape is placed over the blood on the slide should be sucked away by Pasteur
area with its central opening over the aspiration site. pipette, leaving behind marrow particles. If immuno-
4. Skin and periosteum are infiltrated with a local phenotyping or cytogenetic analysis is to be carried
anesthetic. First inject beneath the skin surface and out, further marrow sample should be aspirated in a
advancing the needle further, a larger amount is second syringe and dispensed in a tube containing
injected into the periosteal surface. heparin anticoagulant.
5. After waiting for 5 minutes for anesthesia to take 8. After completion of aspiration, the stylet should be
effect, bone marrow aspiration needle is inserted reinserted into the needle and the needle is removed.
along with the fitted stylet. (Stylet prevents blockage Sterile gauze is placed over the site and light pressure
of lumen of needle by tissues through which needle is applied till bleeding ceases. A larger dressing is
passes). When the bone is reached, the needle is then applied.
rotated clockwise and anticlockwise and slowly
Bone Marrow Trephine Biopsy
advanced into the bone, maintaining steady and firm
pressure. When the marrow is reached, a slight “give” Preparation of the patient and local anesthesia are similar
(decrease in resistance) will be noted. The needle is to aspiration. A short acting intravenous sedative is
advanced for 1-2 mm into the marrow and the stylet preferable in adults. In children, general anesthesia may
is removed. If the needle is placed correctly, it will be be necessary.
fixed by the surrounding bone and will remain rigid Percutaneous trephine biopsy of bone marrow is
and unmoving. commonly obtained from posterior superior iliac spine.

tahir99 - UnitedVRG
vip.persianss.ir
224 Essentials of Clinical Pathology

Jamshidi or Islam needles are commonly used. Often, bone is longer than that for blood films. On smears, marrow
marrow aspiration and biopsy are combined together; particles are dragged towards the tail end of the smear
aspiration is carried out first followed by biopsy. If both and after staining appear as dark blue-violet irregular
aspiration and biopsy are combined, then, after aspiration, granules. Rest of the film stains even pink. Routinely,
either (i) the needle is advanced a little further (1-3 cm) one marrow smear (containing one or more marrow
into the bone, or (ii) the needle is withdrawn and reinserted particles) should also be stained with Perl’s stain for
through the same skin incision but placed at a different assessment of bone marrow storage iron.
site in the bone (about 1 cm away). For biopsy, the needle
Bone marrow aspiration provides following information:
should be advanced through the bone rotating it clockwise
• Assessment of morphology of bone marrow cells.
10 times. The needle should be removed by anticlockwise
• Assessment of nature of hematopoiesis (normal,
rotation. The biopsy should be removed gently from the
dyshematopoiesis)
hub end of the needle by inserting the stylet through the
point of the needle. For adequate assessment, biopsy • Cytogenetic analysis
should measure at least 1.6 cm in length (Fig. 25.4). Biopsy • Immunophenotyping of abnormal cells in leukemias.
should be placed in a fixative solution (either 10% formalin • Cytochemistry for typing of leukemia
or preferably Helly’s fluid). Dressing should be applied to • Iron stain for assessing iron stores and sideroblasts
the site similar to aspiration. • Microbial culture, e.g. for tuberculosis
Along with bone marrow aspiration/biopsy, peri- With marrow aspiration, marrow architecture and
pheral blood smears should be prepared from finger cellular relations cannot be studied.
prick and venous blood should be collected in EDTA
anticoagulant for cell counts. Bone Marrow Trephine Biopsy
If prior marrow aspiration is not satisfactory, imprint
COMPLICATIONS OF BONE MARROW smears should be prepared by gently rolling the marrow
ASPIRATION AND/OR BIOPSY biopsy specimen on a glass slide. These smears are
1. Local infection: This complication, which is more stained with one of the Romanowsky stains for
likely to occur in neutropenic patients, can be cytological evaluation. Biopsy specimen is then fixed in
prevented if strict aseptic precautions are observed. either 10% formalin or Helly’s fluid. (Helly’s fluid
2. Hemorrhage: Serious hemorrhage can occur if (i) consists of potassium dichromate 2.5 gm; mercuric
marrow biopsy is done without adequate replace- chloride 5 gm; 40% formalin 5 ml; and water 100 ml).
ment cover in coagulation disorders, and (ii) great The biopsy is processed (by decalcification, dehydration,
vessels or heart is injured during sternal aspiration. clearing, and embedding) to obtain paraffin wax blocks.
3. Cardiac tamponade or mediastinitis: This is likely if Less than 4 μm thick sections are cut and stained with
posterior sternal plate is penetrated during sternal hematoxylin and eosin stain and reticulin. It is
aspiration. recommended to mount 5 stepwise serial sections on one
slide to increase the chance of detecting small focal
PROCESSING OF MARROW SPECIMENS lesions. In addition, Giemsa stain is also recommended
for easier differentiation between blood and marrow cells
Bone Marrow Aspiration and mast cells. Iron stain is less informative on biopsy
A drop of aspirated marrow sample is put on each of the since iron is usually lost during decalcification.
several glass slides, excess blood is removed by sucking Bone marrow biopsy provides following information:
with a Pasteur pipette, and smears are prepared with a • Cellularity of bone marrow
‘spreader’. The marrow particles are carried just behind • Bone marrow architecture
the spreader and cellular trails are produced while • Bone structure
spreading. The smears are allowed to dry in the air and • Marrow fibrosis
are labeled. They are stained with one of the • Focal lesions (granulomas, metastatic deposits,
Romanowsky stains. The staining time for marrow films infiltration by lymphoma).

tahir99 - UnitedVRG
vip.persianss.ir
Examination of Bone Marrow 225

Table 25.1: Comparison of bone marrow aspiration and biopsy


Parameter Bone marrow aspiration Bone marrow biopsy

1. Site Iliac spine, sternum, tibia, spinous Iliac spine (posterior superior)
process of vertebra
2. Information obtained Morphology, cytochemistry, iron stain, Cellularity, architecture, fibrosis,
immunophenotyping, culture focal lesions, bone structure
3. Main indications Unexplained cytopenia, suspected Repeated dry tap, aplastic anemia,
hematological malignancy myelofibrosis, focal lesions, hairy cell
leukemia, staging of lymphoma
4. Needle used Salah, Klima Jamshidi
commonly
5. Studies done Romanowsky stain, Iron stain, H and E stain, reticulin stain,
cytochemistry, cytogenetic or immunohistochemistry
molecular genetic analysis,
immunophenotyping, culture
6. Time for routine Same day Up to 7 days
examination report

With marrow biopsy, detailed morphologic assessment • Plasma cell series


of cells is often not satisfactory. Because of decalcification, • Abnormal cells: blasts, carcinoma cells, and necrotic
paraffin wax- embedded sections show cellular distortion cells.
and shrinkage. Plastic embedding of marrow biopsy is • Parasites: malaria parasites, microfilaria, Leishmania
advocated to obtain superior cytologic details (as donovani, and Histoplasma.
decalcification step is not required). • Iron content of marrow (on iron stain).
Comparison of bone marrow aspiration and biopsy
Romanowsky-stained smears are first examined
is presented in Table 25.1.
under low power objective (×10) to assess:
• Cellularity of marrow particles
EXAMINATION OF MARROW SPECIMENS
• Number of megakaryocytes
Bone Marrow Aspiration Smears • Focal metastatic deposits
• Cell distribution and selection of suitable area for
Peripheral blood smear in conjunction with routine
detailed cytologic examination.
hemogram (hemoglobin and cell counts) should be
Assessment of cellularity is based on examination of
examined first before assessing bone marrow smears.
several marrow particles. Cellularity refers to the
Findings should be interpreted in the light of clinical
proportion of hematopoietic cells as compared to the fat
features and relevant laboratory data.
cells in a marrow particle. Cellularity can also be assessed
Examination of marrow smear consists of assessment by examining the density of hematopoietic cells in
of following features: cellular trails behind the marrow particles. Cellularity
• Cellularity can be expressed either in percentage or stating whether
• Differential count marrow particles are normocellular, hypercellular, or
• Myeloid:erythroid ratio hypocellular for age.
• Erythroid series: maturation sequence, type of Megakaryocytes are found in the tail of the smear or
maturation (normoblastic, micronormoblastic, near the marrow particles. About 1-3 megakaryocytes
megaloblastic), cytologic abnormalities. are seen normally per low power field.
• Myeloid series: maturation sequence, cytologic Cellular trails are examined under high power (×40)
abnormalities. and oil immersion objective (×100) for differential count
• Megakaryocyte series: number, abnormal forms. and assessing erythroid and myeloid maturation. For
• Lymphocyte series differential count, at least 500 cells should be counted in
226 Essentials of Clinical Pathology

cellular trails of particles. Normal differential count in necrotic cells, or metastatic cells should be carried out.
bone marrow in adults is shown at the end of this chapter Parasites like Leishmania donovani and Histoplasma
under “Reference Ranges”. capsulatum are detected in macrophages.
Usually a 5-finger differential count is sufficient Routinely, Perl’s stain for iron is done on a marrow
which consists of counting relative percentages of smear (containing at least one marrow particle) to assess
erythroid cells, myeloid cells, lymphoid cells, plasma storage iron and number and types of sideroblasts.
cells, and blast cells. In acute leukemia, myelodysplastic In acute leukemia, cytochemical stains like myelo-
syndrome, lymphoma infiltration, and plasma cell peroxidase, nonspecific esterase, and periodic acid shiff
dyscrasias, a more detailed differential count is essential. are done for typing of acute leukemia.
Myeloid:erythroid (M:E) ratio is the ratio of all
granulocytes and their precursors to all erythroid Iron Staining of Bone Marrow Aspiration Smears
precursor cells. Normal M:E ratio ranges from 2:1 to 4:1. Presence of iron in marrow aspiration smears can be
On an average, there are 3 myeloid precursors for every demonstrated by Perls’ Prussian blue reaction. In this
erythroid precursor. Since M:E ratio is relative it should method, ionic iron reacts with acid ferrrocyanide solution
be interpreted carefully. Increased M:E ratio is observed to form blue-colored ferric ferrocyanide. Iron appears as
when myeloid series is hyperplastic as in infections and bright blue granular aggregates. Perls’ stain does not
myeloproliferative disorders (chronic or acute myeloid detect heme iron of hemoglobin.
leukemia) or when erythroid series is suppressed as in Iron staining is a valuable test for detection of iron
aplastic crisis of hemolytic anemia. Reduced M:E ratio is deficiency and must be carried out as a routine on all
observed when myeloid series is depressed and in aspiration smears (Fig. 25.5). Bone marrow aspiration
erythroid hyperplasia (e.g. hemolytic anemia). smears containing at least one marrow particle are
Maturation of erythroid and myeloid series should required for demonstration of storage iron in macro-
be assessed. Nature of erythroid maturation may be phages. Bone marrow biopsy sections are not suitable
normoblastic, micronormoblastic, or megaloblastic. for evaluation of iron stores since some iron is lost during
Dyshematopoietic features may be suggestive of processing (decalcification step) of tissue.
myelodysplastic syndrome or congenital dyserythro- On marrow smears, stainable iron can be demons-
poietic anemia. trated in siderocytes, sideroblasts, and macrophages of
Detailed cytological evaluation of abnormal cells like reticuloendothelial system. Siderocytes are immature,
blast cells, lymphoma cells, myeloma cells, storage cells, non-nucleated red cells, which contain 1-2 granules of

Fig. 25.5: Iron stain on bone marrow aspiration smear.


Left part of the figure shows blue stained iron granules. Right part of the figure shows no stainable iron
Examination of Bone Marrow 227

non-heme iron. On Romanowsky-stained smears, these Cellularity (Fig. 25.6) varies with the age of the patient.
are called as Pappenheimer bodies. Erythroblasts There is a gradual reduction in red marrow (hematopoietic
containing aggregates of iron are called as sideroblasts. tissue) and proportionate increase in fat cells as age
They are of three types—I, II, and III. In type I sideroblasts advances. A rough estimate of normal cellularity is
(which are seen normally), iron granules are small and obtained by deducting 1 for each year of age from 100 and
few (1-4) in number. Normally, about 25-50% of expressing it as a percentage; expected normal cellularity
erythroblasts contain 1-4, small iron granules. Types II is ±10% of this value. For example, if age of the patient is
and III sideroblasts are abnormal. In type II sideroblasts, 25 years, then expected normal cellularity is 65% to 85%.
iron granules are large and numerous. Type II sidero- The cellularity in the first decade of life is about 80% while
blasts are seen in hemolytic anemia and iron overload. in adults it is about 50%. In older age (>70 years), cellularity
In type III sideroblasts, iron granules are distributed in decreases to about 30%. Also, normally, the cellularity
the form of a ring around the nucleus. The ‘ringed’ immediately beneath the subcortical area is low as
sideroblasts are seen in sideroblastic anemias. compared to the deeper medullary areas.
Iron granules in macrophages of reticuloendothelial Normally bone marrow biopsy shows bony trabecule
system represent storage form of iron. Normally, a few,
(lamellar bone) separated by interconnecting spaces
small granules are seen. In iron deficiency anemia, there
is complete lack of stainable iron in the erythroblasts and containing bone marrow. The bony trabecule are thin,
macrophages. Lack of stainable iron in erythroblasts and irregular, and show osteocytes within lacune. The
increased amount of iron in macrophages is a feature of trabecule show lining of endosteum (osteoblasts and
anemia of chronic disease. Marked increase of storage iron osteocytes). In a disease called osteopetrosis, bony
in macrophages occurs in thalassemias and iron overload. trabecule are markedly thick with severe reduction in
marrow space; there are cartilaginous plates in trabecular
Bone Marrow Trephine Biopsy
bone; and numerous osteoclasts are present along the
Hematoxylin and eosin-stained sections of marrow endosteal surface.
trephine biopsy are initially examined under low power High power examination is carried out to assess relative
objective to assess cellularity, to identify focal lesions like
proportion of myeloid and erythroid cells, location of
granuloma, lymphoma infiltrates and clumps of metastatic
malignancy, and to assess bone structure and number of hematopoietic precursors, pattern of infiltration of
megakaryocytes. Because of variable cellularity between lymphoma cells, morphology of abnormal cells, and
particles and dilution with peripheral blood in marrow parasites.
aspiration smears, assessment of cellularity is more Normally, erythroid precursors are located in clusters
reliably performed on biopsy sections. in the center of marrow spaces, while granulocyte

Fig. 25.6: Cellularity of bone marrow on biopsy: (A) Normocellular; (B) Hypercellular; and (C) Hypocellular
228 Essentials of Clinical Pathology

precursors are present close to the trabecule. In REFERENCE RANGES


myelodysplastic syndrome, clusters of myeloblasts and
Differential Count in Bone Marrow in Adults
promyelocytes are located in central marrow cavity; this
is called as abnormal localization of immature precursors • Myeloblasts: 0-3%
or ALIP. • Promelocytes: 2-5%
• Neutrophil myelocytes: 8-15%
In infiltration of bone marrow by non-Hodgkin’s
• Metamyelocytes: 9-24%
lymphoma, pattern of infiltration may be paratrabecular, • Neutrophils (including band forms): 14-26%
focal non-paratrabecular, interstitial, and diffuse. • Erythroblasts: 15-36%
Similarly, distinct patterns of infiltration are found in • Lymphocytes: 5-20%
chronic lymphocytic leukemia like diffuse, interstitial, • Plasma cells: 0-3%
nodular, or mixed (nodular and interstitial) which has • Myeloid:erythroid (M:E) ratio: 2:1 to 4:1.
prognostic significance. Iron Staining of Bone Marrow Smears
In idiopathic myelofibrosis, collagen deposition can
• Sideroblasts 30-50% of all erythroblasts
be made out readily on H and E stained sections. For • No ringed sideroblasts
demonstration of reticulin fibers, a silver impregnation
technique is required. Reticulin fibers are increased in CRITICAL VALUES
acute leukemias, chronic myeloid leukemia, poly- • New diagnosis of a hematological malignancy or
cythemia vera, myelofibrosis, hairy cell leukemia, aplastic anemia or severe thrombocytopenia
metastatic carcinoma, and certain inflammatory • Parasites.
conditions. BIBLIOGRAPHY
If necessary, special stains like Ziehl-Neelsen stain,
1. Bain BJ. Bone marrow aspiration. J Clin Pathol
Giemsa stain, Congo red stain, stains for fungi, and 2001;54:657-63.
immunoperoxidase stains can be applied. 2. Bain BJ. Bone marrow trephine biopsy. J Clin Pathol
During decalcification of bone marrow biopsy 2001;54:737-42.
3. Hoffbrand AV, Moss PAH, Pettit JE (Eds). Essential
specimen for paraffin wax embedding, many cellular Hematology. (5th Ed). Blackwell Publishing Ltd, 2006.
enzymes are destroyed and therefore cytochemical stains 4. Hyun BH, Gulati GL, Ashton JK. Bone marrow exami-
(for typing of leukemias), which depend on enzymatic nation: techniques and interpretation. Hematology/
Oncology Clinics of North America 1988;4:513-52.
reactions within cells, cannot be applied. For plastic
5. Knowles S and Hoff brand AV. Bone marrow aspiration
embedding of tissues, decalcification is not required and and trephine biopsy. (1) BMJ 1980;281:204-5. (2) BMJ
therefore cytochemistry is possible. 1981;281:280-1.
26

Diagnosis of Malaria and


Other Parasites in Blood

MALARIA LIFE CYCLE OF MALARIA PARASITES


Malaria is endemic in tropical and subtropical develo-
Life history of malaria parasite consists of two cycles of
ping countries where it is a major public health problem.
development: asexual cycle or schizogony that occurs in
Every year there are about 300-500 million clinical cases
of malaria with 1-2 million deaths, most of them in Africa. humans and sexual cycle or sporogony that occurs in
Inadequate administration and implementation of mosquitoes (Fig. 26.1).
malaria control measures, failure of insecticides, and 1. Asexual cycle (human cycle, schizogony): This
emergence of drug-resistance strains of the parasite are occurs in the liver cells and red blood cells of infected
responsible for resurgence of malaria. humans, and therefore humans are the intermediate
There are four species of malaria parasites that infect
hosts of the malaria parasite (Schizogony refers to the
humans: Plasmodium vivax, P. falciparum, P. malariae, and
process of reproduction in protozoa in which there is
P. ovale. P. vivax is the most widely distributed species in
the world. In India, the prevalent forms of infections are: production of daughter cells by fission). The human
P. vivax (70%), P. falciparum (25-30%), mixed P. vivax and cycle begins when infected female Anopheles
P. falciparum (4-8%), and P. malariae (< 1%). P. malariae is mosquito bites a person and sporozoites are injected
localised to certain foci, especially in some areas of into the circulation. There are four stages of human
Karnataka. P. ovale is mainly restricted to West Africa. cycle.

Fig. 26.1: Life cycle of malaria parasite


230 Essentials of Clinical Pathology

a. Pre-erythrocytic schizogony (Hepatic schizogony): P. vivax, P. ovale, and P. malariae complete the
Inoculated sporozoites rapidly leave the circulation erythrocyte schizogony in general circulation.
to enter the liver cells where they develop into Schizonts of P. falciparum induce membrane
hepatic (pre-erythrocytic) schizonts (Schizonts are changes in red cells, which causes them to adhere
cells undergoing schizogony). One sporozoite to the capillary endothelial cells (cytoadherence).
produces one tissue form. Hepatic schizonts Therefore, in P. falciparum infection, erythrocyte
rupture to release numerous merozoites in schizogony is completed in capillaries of internal
circulation (Merozoites are daughter cells organs and usually only ring forms are seen in
produced after schizogony). Up to 40,000 merozoi- circulation.
tes are produced in the hepatic schizont. c. Gametogony: After several cycles of erythrocytic
In P. falciparum infection, all of the hepatic schizogony, some merozoites, instead of develo-
schizonts mature and rupture simultaneously; ping into trophozoites and schizonts, transform
dormant forms do not persist in hepatocytes. In into male and female gametocytes. These sexual
contrast, some of the sporozoites of P. vivax and forms are infective to mosquito and the person
P. ovale remain dormant after entering liver cells harboring them is called as a “carrier”. Gameto-
and develop into schizonts after some delay. Such cytes are not pathogenic for humans.
persistent forms are called as hypnozoites; they d. Exoerythrocytic schizogony: In P. vivax and P.
develop into schizonts at a later date and are a ovale infections, some of the sporozoites in liver
cause of relapse. cells persist and remain dormant. These dormant
b. Erythrocytic schizogony: Merozoites released forms in liver cells are called as hypnozoites. They
from rupture of hepatic schizonts enter the red become active and develop into schizonts a few
days, months, or even years later. These schizonts
blood cells via specific surface receptors. These
rupture, release merozoites, and cause relapse.
merozoites become trophozoites that utilize red
Exoerythrocytic schizogony is absent in P.
cell contents for their metabolism. A brown-black
falciparum infection and therefore relapse does not
granular pigment (malaria pigment or hemozoin)
occur. Hence, P. vivax and P. ovale are called as
is produced due to breakdown of hemoglobin by
relapsing plasmodia while P. falciparum and
malaria parasites. The fully formed trophozoite
P. malariae are known as non-relapsing plasmodia.
develops into a schizont by multiple nuclear and
2. Sexual cycle (mosquito cycle, sporogony): The sexual
cytoplasmic divisions. Mature schizonts rupture
cycle begins when a female Anopheles mosquito
to release merozoites, red cell contents, malarial
ingests mature male and female gametocytes during
toxins, and malarial pigment. (This pigment is
a blood meal. First, 4-8 microgametes are produced
taken up by monocytes in peripheral blood and from one male gametocyte (microgametocyte) in the
by macrophages of reticulo-endothelial system. In stomach of the mosquito; this is called as ex-
severe cases, organs which are rich in macro- flagellation. The female gametocyte (macrogameto-
phages like spleen, liver, lymph nodes, and bone cyte) undergoes maturation to produce one macro-
marrow become slate-gray or black in color due gamete. By chemotaxis, microgametes are attracted
to hemozoin pigment). Rupture of red cell toward the macrogamete; one of the microgametes
schizonts corresponds with clinical attack of fertilizes the macrogamete to produce a zygote. The
malaria. Released merozoites infect new red cells zygote becomes motile and is called as ookinete.
and enter another erythrocytic schizogony cycle. Ookinete penetrates the lining of the stomach and
This leads to rapid amplification of plasmodia in comes to lie on the outer surface of the stomach where
the red cells of the human host. In P. falciparum, it develops into an oocyst. On further growth and
P. vivax, and P. ovale infections, cycle of schizogony maturation, multiple sporozoites are formed within
lasts for 48 hours, while in P. malarie infection it the oocyst. After complete maturation, oocyst
lasts for 72 hours. Merozoites of P. vivax and P. ruptures to release sporozoites into the body cavity
ovale preferentially invade young red cells or of the mosquito. Most of the sporozoites migrate to
reticulocytes while those of P. falciparum infect red the salivary glands. Infection is transmitted to the
cells of all ages. Senescent red cells are preferred humans by the bite of the mosquito through saliva
by P. malariae. when it takes a blood meal.
Diagnosis of Malaria and Other Parasites in Blood 231

CLINICAL FEATURES appears dark brown or black in color due to hemo-


globinuria.
Clinically malaria is characterized by paroxysms of high-
Infection by P. malariae can cause nephrotic syndrome.
grade fever along with anemia and splenomegaly. There
are three stages of a typical malarial paroxysm: (i) cold
MODES OF TRANSMISSION
stage (lasts for 20 min to 1 hr): There is sudden onset of
fever, shivering, and extreme cold; (ii) hot stage (1-4 hr): Malaria is transmitted by the bite of certain species of
Fever rises to its maximum and there are associated infected female Anopheles mosquitoes. Other rare modes
severe headache and body ache; (iii) sweating stage (2-3 of transmission are blood transfusion, use of conta-
hrs.): Patient perspires profusely and temperature falls. minated syringes or needles, and congenital (from
The duration of each paroxysm is about 6-10 hours and mother to the fetus). Hepatic schizogony does not occur
coincides with the rupture of erythrocytes and release of when malaria is transmitted through transfusion,
showers of merozoites. Febrile paroxysms recur every contaminated syringes, or congenital route; therefore, in
48 hours (in P. vivax, P. falciparum, and P. ovale infections) case of P. vivax infection, relapse will not occur.
that is called as tertian malaria or every 72 hours (in
P.malariae infection) called as quartan malaria. ROLE OF GENETIC FACTORS IN MALARIA
Anemia usually results from destruction of para- 1. Sickle cell trait and β -thalassemia trait: Young
sitized red cells; other causes are suppression of children with sickle cell trait (heterozygotes) develop
erythropoiesis in bone marrow and immune hemolysis. comparatively mild form of falciparum malaria.
Splenomegaly is an important feature and with However, persons with sickle cell anemia (homo-
repeated paroxysms there is a significant enlargement zygous for sickle cell gene) are not protected. Theory
of spleen. Splenomegaly is secondary to activation and of balanced polymorphism proposes that selective
hyperplasia of mononuclear phagocytic system. advantage gained by sickle heterozygotes is balanced
Other clinical features include myalgia, joint pains, by disadvantage of homozygous state. β-thalassemia
thrombocytopenia, and hypoglycemia. trait is also protective against severe falciparum
Relapse is a feature of malaria caused by P. vivax and malaria.
P. ovale and is due to persistence of hypnozoites in liver 2. Glucose-6-phosphate dehydrogenase (G-6-PD)
cells. It may occur months or even years after the initial deficiency: Protection against severe falciparum
attack. Relapse does not occur with P. falciparum infection. malaria is afforded to G-6-PD-deficient female
Untreated P. falciparum infection can be severe and heterozygotes.
fatal. This is because of high degree of parasitemia and 3. Newborns: For the first few months of life, newborn
sequestration of parasitized red cells in capillaries of infant has high levels of hemoglobin F in red cells
internal organs. The red cells containing trophozoites and that suppresses the growth of malaria parasite.
schizonts of P. falciparum express, on their surface, 4. Duffy antigen: The Fy (a-b-) phenotype (i.e. Duffy
positively charged parasite proteins and become ‘sticky’. antigen-negative blood group) in blacks confers
These red cells adhere to each other and to endothelial resistance against P. vivax infection. This is because
cells of capillaries of internal organs (cytoadhesion). This P. vivax parasite enters the red cells at glycophorin
leads to clogging of microcirculation by parasitized red receptors present on Duffy antigen site. P. vivax
cells with resultant rupture of capillaries. Two serious infection is very rare in West Africa due to the lack of
complications of falciparum malaria are cerebral malaria Duffy antigen on red cells of the population.
and blackwater fever. Cerebral malaria clinically
manifests as hyperpyrexia, convulsions, coma, and Methods of Diagnosis
sometimes death. In blackwater fever, there is sudden Malaria can be diagnosed by following methods:
onset of massive intravascular hemolysis (which causes • Clinical diagnosis
hemoglobinemia, hemoglobinuria, and hyperbili- • Microscopic examination of blood smears
rubinemia) and high-grade fever. Renal failure is • Rapid diagnostic tests (Detection of malaria parasite
common. Parasites are not detectable in blood. Urine antigen or enzyme)
232 Essentials of Clinical Pathology

• Fluorescence microscopy After complete drying, thick smear is stained with a


• Serologic techniques Giemsa stain or a Field’s stain. Red cells are lysed during
• Detection of nucleic acid of parasite by polymerase staining and parasites are better visualised against a clear
chain reaction. background. Thick smear (since it allows examination
1. Clinical diagnosis: This is the most common method of a relatively large amount of blood) is more sensitive
for diagnosis of malaria in endemic countries. than thin smear for detection of malaria parasites,
However, this method is unreliable since symptoms particularly if they are few in number. For thick smear, a
of malaria overlap with those of many other febrile large drop of blood is collected in the center of glass slide.
illnesses like viral fever, viral encephalitis, and It is spread with the corner of a spreader slide or a stick
typhoid fever. in such a manner that an evenly spread circular or
2. Microscopic examination of thick and thin smears: rectangular smear of size 15 × 15 mm is obtained. After
The most commonly used, fairly rapid, and accepted labeling, smears are allowed to dry in the air. For better
method for diagnosis of malaria is microscopic results, thick smear can be dried in an incubator at 37°C
examination of stained blood smears. It remains the for 15 min. Thick smear should not be fixed since it is to
gold standard for diagnosis of malaria. Blood smear be dehemoglobinised.
should be obtained before the next expected febrile Thin smear is prepared as outlined in Chapter 22
paroxysm or at the onset of fever and chills. Collection (Blood Smear). It is air-dried, fixed with methanol, and
of blood immediately following a paroxysm of fever stained with a Giemsa stain or Leishman stain. Thin
is not likely to show intrerythrocytic parasites because smear is used for definitive identification of parasite
of lysis of parasitized red cells. Blood sample should species, determination of parasite density (if parasitemia
be taken before antimalarial drugs are given. After is very high), and for detection of any associated
administration of antimalarial drugs, parasites morphologic abnormalities of blood cells. It is fixed with
become scarce in blood and alter in morphology. methanol for 1-2 minutes before staining.
Ideally, blood should be collected by skin puncture Thick and thin smears can also be prepared on the
and smears made immediately. Adhesion of blood to the same slide.
slide and subsequent staining are affected if antico- It is recommended to examine at least 100 oil
agulated blood sample is used. If it is necessary to use immersion fields in thick smear and 200 oil immersion
anticoagulated blood, blood should be collected in EDTA fields in thin smear before reporting the smear as negative
and smear made as early as possible; delay causes for malaria parasite.
morphologic changes in parasites, which may make If blood smear is negative for malaria parasite despite
diagnosis difficult. strong clinical suspicion, repeat smears should be taken.
One thick smear (for detection of parasites) and one A buffy coat preparation can also be helpful in such a
thin smear (for definitive identification of species) should case. EDTA-anticoagulated blood is centrifuged and a
be prepared (Fig. 26.2). smear is prepared from buffy coat layer and from red
cells just below it. Parasitized red cells get concentrated
just beneath the buffy coat layer.
In P. vivax infection, almost all red cell stages of
parasite are represented in the blood smear. In P.
falciparum infection, usually only ring forms or gameto-
cytes are seen. Presence of mature trophozoites and
schizonts of P. falciparum in blood smear are indicative
of a serious infection. If only gametocytes of P. falciparum
are seen in untreated subjects, it denotes active,
suppressed infection; if seen in treated subjects, it carries
no significance.
Identification of malaria parasites: Different stages of
malaria parasites are found in peripheral blood. These
Fig. 26.2: Thick and thin blood smears (unstained) are trophozoites, schizonts, and gametocytes. The four
Diagnosis of Malaria and Other Parasites in Blood 233

species of malaria parasites can be distinguished graphic tests. Also, in some cases of falciparum malaria,
morphologically by microscopic examination of thin parasites get sequestered in the capillaries of internal
blood smear. organs and are not detectable on blood smears (but can
Microscopic examination of blood smears for be detected by immunochromatographic tests).
diagnosis of malaria is a sensitive test; it can detect about If P. falciparum is identified, it is necessary to estimate
50 parasites/μl in experienced hands. parasite density. Since gametocytes are non-pathogenic,
Morphological features of malaria parasites are they should be excluded while counting parasites. Two
summarized and compared in Table 26.1. Stages of P. methods for estimation of parasite density are given
vivax and P.falciparum are shown in Figures 26.3 and 26.5. below.
Blood smears in P. vivax and P. falciparum are shown in 1. In a thick smear, number of parasites is counted till
Figures 26.4 and 26.6 respectively. 200 white blood cells are observed. Number of
As compared to the recently available rapid immuno- parasites present per μl of blood is calculated from
chromatographic tests (see below), microscopic examina- the following formula:
tion of the blood smear is relatively inexpensive.
Number of parasites
However, it needs good quality control, is labor- Total leukocyte count/µl × ———————————
intensive, and takes more time than immunochromato- 200

Table 26.1: Morphological comparison of Plasmodia

Plasmodium falciparum
Often only ring forms and gametocytes are seen; multiple rings in a single cell common; red cell size normal; high
parasite density.

Early trophozoite Late trophozoite Schizont Gametocyte


A delicate, small Compact blue ring with Very rarely seen except Crescentic or banana-
uniformly fine cytoplasmic 1-2 red chromatin dots in cerebral malaria; shaped; larger than a red
ring with 1-2 small chromatin contains 18-32 cell
dots; ring may be attached to merozoites which fill
the red cell margin (accole form). 2/3rds of a red cell; a
single block of dark
brown pigment

Plasmodium vivax
Often all stages are seen; single parasite in a red cell; red cell enlarged and shows Schuffner’s dots (fine, pink
granules); medium parasite density.

Blue cytoplasmic ring Irregularly thick 12-24 merozoites Large and spherical
1/3rd the diameter of the cytoplasmic ring arranged rosette-like
red cell; one side of ring (ameboid); large granular yellow-brown
thicker; red chromatin at red chromatin dot pigment in center
thinner part of ring

Plasmodium ovale
Often all stages are seen; single parasite in a red cell; red cell slightly enlarged, oval, and with fimbriated margins;
prominent Schuffner’s dots.
Thick, dense blue ring Compact ring; slightly 8-12 irregularly placed Oval; prominent
with a large red amoeboid merozoites Schuffner’s dots
chromatin dot

Plasmodium malariae
Often all stages are seen; single parasite in a red cell; red cell normal in size; no Schuffner’s dots; low parasite
density.
Similar to P. vivax Compact band-like ring 6-12 merozoites in a Round or oval
across the red cell “daisy-head” pattern
•••••••••••••••••

234 Essentials of Clinical Pathology

Fig. 26.3: Stages of P. vivax

2. In a thin smear, number of parasites amongst 1000


red cells is counted and reported as a percentage.
Number of parasites in 1 μl of blood can be calculated
if red cell count in millions/μl is known; if it is not
known, then it can be arbitrarily taken as 5 million/
μl.

Number of parasites in 1 μl of blood = Red cell count in


million/cmm × parasite percentage

Estimation of degree of parasitemia is of significance


in P. falciparum infection. Percentage parasitemia
exceeding 10% is an indication for exchange transfusion.
In patients taking antimalarial treatment, percent
parasitemia should be obtained daily till no more
parasites (excluding gametocytes) are detectable.

Reporting the result: In the presence of malaria parasite,


Fig. 26.4: P. vivax infection is associated with
blood smear should be reported as follows:
simultaneous presence of multiple stages • Blood smear is positive for malaria parasite
• Name of species
Usually, total leukocyte count is taken as 8000/ • Red cell stages seen
μl and the number of parasites is multiplied by 40 to • Parasite density (in case of P. falciparum)
get the result. However, if accurate leukocyte count Comparison of morphology of P. vivax and
is known, then a better estimate of parasite density is P. falciparum is shown in Figure 26.7.
obtained. 3. Detection of malaria parasite antigen by rapid
To obtain percent parasitemia, figure of number immunochromatographic tests: In recent years,
of parasites amongst 200 white blood cells is divided commercial manufacturers have introduced simple
by 1250. non-microscopic rapid diagnostic tests. These tests

Fig. 26.5: Stages of P. falciparum


Diagnosis of Malaria and Other Parasites in Blood 235

Many commercial test kits are available with differing


test procedures. General principle of these tests is as
follows. Blood sample is collected by skin puncture. It is
then mixed with a buffer solution provided with the kit;
this solution causes lysis of red cells and also contains a
specific antibody labeled with a dye. If the antigen under
investigation is present in blood, formation of antigen-
antibody complex occurs. The labeled antigen-antibody
complex is then allowed to migrate along a nitrocellulose
test strip by capillary action (The manufacturer has
impregnated this test strip with a monoclonal antibody
against the antigen across the strip. The strip also contains
a line of positive control to check whether the test has
been performed correctly and whether the reagents are
functional). In the last step, a washing solution is added
to remove hemoglobin from lysed red cells and to
visualise the reaction. If the blood sample contains the
antigen under investigation, the labeled antigen-
Fig. 26.6: Blood smear in P. falciparum infection showing ring
forms.Usually a single stage is seen in P. falciparum infection antibody complex will form, it will migrate up the strip,
and will be captured by the pre-deposited monoclonal
antibody; this results in the appearance of a colored line
detect antigens of malaria parasites by immuno- across the strip.
chromatographic method. The antigens against Tests that detect P. falciparum, P. vivax, and other
which the commercial test kits are currently available species are available. Tests which detect HRP-2 antigen
are: of P. falciparum have sensitivity and specificity of >90%
• Histidine rich protein-2 (HRP-2) synthesized by (if parasite density is greater than 100/μl).
asexual blood stages and young gametocytes of
P. falciparum and expressed on red cell surface. Commercial test kits can be helpful in following
• Parasite lactate dehydrogenase (pLDH), an situations:
enzyme of glycolytic pathway found in all four • Confirmation of P. falciparum infection if there is
human malaria species; there are different forms uncertainty about the identity of malaria parasite on
of pLDH for each species. Level of pLDH blood smear.
correlates with parasite density. • Confirmation of P. falciparum in mixed infections.

Fig. 26.7: Comparison of P. falciparum and P. vivax morphology


236 Essentials of Clinical Pathology

• Diagnosis of P. falciparum in endemic remote Acridine orange stains all the cells that contain
peripheral areas where properly trained technicians nucleic acids. Therefore, considerable experience
and facilities for microscopic diagnosis are not is required to distinguish fluorescing parasites
available. Local health workers with little training can from other cells containing nucleic acids. This
perform rapid diagnostic tests. method also needs expensive apparatus and
• Rapid, self-diagnosis of malaria by tourists traveling materials. Howell-Jolly bodies (nuclear remnants
to endemic countries. Some manufacturers have in red cells in certain anemias) give a false positive
developed self-help diagnostic kits for travelers. reaction. Schizonts and gametocytes get localized
Positive rapid diagnostic tests (for HRP-2) persist for in the buffy layer and are missed. Specific
several days following successful treatment of P. identification of species is difficult and it is
falciparum infection (due to persistence of antigens in necessary to use Romanowsky-stained smears for
blood). Therefore these tests are not useful in following this purpose. Despite these disadvantages, this
response to treatment. Also these tests are not able to technique is a more rapid alternative to blood
estimate parasite density. Distinction between non- smears.
pathogenic gametocyte stage and other pathogenic stages b. Kawamoto technique: Blood smears are prepared,
is not possible. stained with a fluorescent dye (acridine orange),
Comparison between blood smear and rapid diag- and examined by fluorescence microscopy. An
nostic tests is given in Table 26.2. interference filter designed for acridine orange is
4. Fluorescence microscopy: There are two fluorescence placed in the pathway of transmitted light beam
microscopy techniques employed for diagnosis of and a barrier filter is placed in the eyepiece. Nuclei
malaria: quantitative buffy coat (QBC) system (Becton of malaria parasites fluoresce bright green and
Dickinson), and Kawamoto technique. cytoplasm red. Nuclei of white cells also fluoresce
a. Quantitative Buffy Coat (QBC) system (Becton bright green. Definitive species identification is
Dickinson): Blood is centrifuged in a special difficult; therefore Romanowsky-stained smears
capillary tube which contains a float and which is are required for species diagnosis. Kawamoto
coated with acridine orange (a fluorescent dye) technique is less expensive than QBC system.
and an anticoagulant. Following centrifugation, 5. Detection of nucleic acid sequences of malaria
malaria parasites get concentrated in the upper parasites: Malaria parasites can be detected by
layer of red cells just below the buffy coat and are identification of specific nucleic acid sequences in
stained with the fluorescent dye. When the their DNA. Methods based on polymerase chain
capillary tube is viewed using a special objective reaction (PCR) have been developed for identification
(paralens) attached to the fluorescence micro- of DNA of malaria parasite. Species diagnosis is also
scope, malaria parasites fluoresce green yellow possible. PCR-based methods can detect very low
against a dark red-black background. Nuclei of levels of parasites in blood (< 5 parasites/μl of blood)
trophozoites fluoresce bright green. with very high sensitivity and specificity.

Table 26.2: Comparison of blood smear and commercial rapid diagnostic tests for malaria diagnosis
Parameter Blood smear Rapid diagnostic tests
1. Sensitivity 5-10 parasites/μl 40-100 parasites/μl
2. Species identified All Depends on test kit
3. Parasite density Can be estimated Cannot be estimated
4. Performance Laborious Easy
5. Time required 1 hour 15-20 minutes
6. Detection of sequestered P. falciparum No Yes
7.Distinction between gametocytes and other stages Yes No
8. Cost Low High
Diagnosis of Malaria and Other Parasites in Blood 237

Molecular methods can be useful in the diagnosis


of malaria, in following response to treatment, in
epidemiological surveys, and for screening of blood
donors. They can also be used as a standard to judge
other methods of malaria diagnosis. However, these
methods cannot be routinely applied because of the
high cost, need for special equipments and materials,
and lengthy procedure (24 hours). Presently they can
be used as a research tool in malaria control programs,
and to carry out quality control checks on microscopic
diagnosis.
6. Serologic methods: Indirect immunofluorescence or
hemagglutination tests are available which detect
antibodies against malaria parasites in patient’s Fig. 26.8: Life cycle of Wuchereria bancrofti
serum. These tests are helpful for: (i) retrospective
epidemiological surveys in endemic areas, (ii)
screening of blood donors for asymptomatic infec- After fertilization, female worms produce numerous
tion, and (iii) retrospective diagnosis. However, these microfilariae, which migrate into venous blood (via large
tests are not helpful for routine diagnosis of active lymphatic ducts) and reach arterial circulation through
infection as they cannot distinguish between current pulmonary capillaries. The adult worms inhabit the
and past infection. Thus they cannot guide treatment. lymphatics and live for many (5-10) years.
The microfilariae are ingested by the mosquitoe when
LYMPHATIC FILARIASIS it takes a blood meal. Microfilariae cast off their sheaths
in the stomach of the mosquitoe, penetrate the gut wall,
Lymphatic filariasis is a parasitic infection caused by
and migrate into the thoracic muscles where they reside
nematode worms Wuchereria bancrofti, Brugia malayi, and
and grow into infective larvae. After about 2 weeks of
Brugia timori. Lymphatic filariasis affects more than 120 development, the mature infective forms migrate to the
million people in the tropical and subtropical countries. mouthparts of the mosquitoe and are transmitted to the
In India, lymphatic filariasis is caused by Wuchereria definitive host through the bite.
bancrofti (majority of cases) and Brugia malayi (mainly in
southwest India). (Brugia timori is restricted to Indonesia). CLINICAL FEATURES
According to World Health Organization, one-third of Clinical features are variable. Also, after acquisition,
the people affected by lymphatic filariasis are in India. infection may take years to manifest clinically. Many
The prevalence of this disease appears to be on the rise infected individuals do not exhibit any clinical manifes-
due mainly to proliferation of slums and expansion of tations even though they have numerous circulating
breeding sites for mosquitoes. microfilariae and harbor adult worms in lymphatics.
In acute disease, repeated attacks of fever are
Life Cycle associated with lymphangitis (filarial fever). Infla-
mmation occurs in lymphatics of limbs, genitals
Filarial worms pass their life cycle in two hosts: humans
(epididymis, spermatic cord, and vulva), and breasts.
(definitive host) and mosquitoes (intermediate host)
Lymphangitis is often associated with enlargement of
(Fig. 26.8).
regional lymph nodes (inguinal or axillary). Repeated
The disease is transmitted to the humans by the bite episodes of lymphangitis cause fibrosis and obstruction
of infected female mosquitoes of the genera Culex, of lymphatics with accumulation of lymph fluid in
Anopheles, or Aedes. The infective larvae are deposited surrounding tissues (lymphedema).
on the human skin when the mosquito takes a blood The late manifestations of filariasis are hydrocele and
meal. The larvae then enter the host through the puncture elephantiasis. In hydrocele, sac surrounding the testis is
wound. The larvae reach the lymphatic channels and filled with fluid. In elephantiasis, there is extreme
mature into adult male and female worms (5-18 months). edematous enlargement of the affected part (limbs,
238 Essentials of Clinical Pathology

external genitals, or breasts) with coarse thickening and prepared from capillary blood collected at the appropriate
fissuring of skin. Local bacterial and fungal infections time, and if clinical suspicion is strong, concentration
are common. Microfilariae are usually not found in blood techniques are employed. This is because circulating
of patients with hydrocele and elephantiasis. microfilariae are often scanty and sensitivity of
In some cases, there is passage of milky-white chyle microfilarial detection increases when volume of blood
in urine (chyluria). This is due to rupture of distended sampled is increased.
urogenital lymphatics, which are connected to the
intestinal lymphatic vessels carrying chyle. Microscopic Concentration techniques
examination of such urine shows microfilariae. Membrane filtration: This is a sensitive method but is
expensive for routine use in endemic areas. Anticoa-
gulated venous blood (10 ml) is passed through a
LABORATORY DIAGNOSIS
polycarbonate membrane filter of 3 μm or 5 μm pore size.
Diagnostic methods in lymphatic filariasis include: Following this 10 ml of methylene blue saline solution is
• Microscopic examination of blood for demonstration passed through the filter for staining the microfilariae.
of microfilaria Microfilariae are trapped and retained on the filter, which
• Detection of filarial antigen in blood by rapid is placed on a glass slide and examined under the
immunochromatographic test microscope.
• Demonstration of microfilariae in hydrocele fluid or Microhematocrit tube or capillary tube method: Two
chylous urine heparinised capillary tubes are filled with blood from
• Demonstration of adult worms in lymph node biopsy skin punctures (or two plain capillary tubes are filled
• Detection of parasite DNA by polymerase chain with anticoagulated venous blood). After sealing the dry
reaction. ends with a suitable sealant, tubes are centrifuged in a
microhematocrit centrifuge for about 5 minutes. The
Microscopic Examination of Blood for capillary tubes are placed on a glass slide and fixed with
Demonstration of Microfilaria adhesive tape. Plasma just above the buffy coat layer is
examined for motile microfilariae under the microscope
Since some species exhibit periodicity (i.e. circulation of
(Fig. 26.9).
microfilariae in increased numbers at certain times of the
day), blood should be collected at the correct time to Lysed venous blood method: 10 ml of venous blood is lysed
improve the chances of detection. For Wuchereria bancrofti by saponin-saline solution. The hemolysate is centri-
and Brugia malayi showing nocturnal periodicity, blood fuged, supernatant is discarded, and the sediment is
should be collected at night between 10 p.m. to 4 a.m. placed on glass slide. After adding a drop of methylene
Microfilariae are present in greater numbers in capillary blue solution, a coverslip is placed, and the preparation
blood than in venous blood; therefore, skin puncture is is examined under the microscope for microfilariae.
preferred. Usually microfilariae are scanty in peripheral
blood so that concentration techniques may be necessary Lysed capillary blood method: 0.1 ml of blood obtained by
for their demonstration. skin puncture is added to 1 ml of saponin-saline solution
Following microscopic methods can be used for to cause lysis of red cells. After centrifugation, super-
detection of microfilariae in peripheral blood: natant is discarded and sediment is placed on a glass
• Thick blood smear slide. A drop of methylene blue solution is added and a
• Concentration techniques: membrane filtration, coverslip is placed over it. The entire preparation is
microhematocrit centrifugation, lysed venous blood examined under the microscope for motile microfilariae.
technique, lysed capillary blood technique.
Morphology of Microfilariae on
Thick Blood Smear Romanowsky-stained Blood Smears
A thick blood smear is spread from 20 μl of capillary blood Wuchereria bancrofti: Microfilariae measure about 300 μ in
on a glass slide, air-dried, and stained with a Romanowsky length and 8 μ in breadth. They have a hyaline sheath,
stain. If microfilariae are not detected in thick smears which stains pink. There are distinct nuclei in the central
Diagnosis of Malaria and Other Parasites in Blood 239

Fig. 26.9: Microhematocrit tube concentration technique for demonstration of microfilaria.

shows two distinct nuclei. Cephalic space is twice as long


as it is broad. Instead of smooth curves to the body, there
are kinks.

Detection of Filarial Antigen in Blood by


Rapid Immunochromatographic Test
A highly sensitive and specific test for diagnosis of active
infection by Wuchereria bancrofti is ICT Filariasis Card Test
(ICT Diagnostics), which is available commercially. This
test detects circulating antigens of this organism in a
fingrprick blood sample of infected individuals. In
contrast to the microscopic tests, blood sample can be
collected at any time of the day. There is no cross-
reactivity with other filarial organisms like Brugia malayi.
Positive result is obtained even if microfilariae are not
circulating and live adult worms are present in the
lymphatics. The test remains positive for up to 18 months
following successful therapy of filariasis. Apart from
Fig. 26.10: Microfilaria of W. bancrofti in blood smear diagnosis of individual patients, this test can be applied
to assess the prevalence of filarial infection in a
axis of the body. Nuclei are not present in the tip of the population. Time required for the test is about 10-15
tail. Cephalic space (present at the anterior end) is as long minutes.
Other tests have been developed for detection of
as it is broad. Tip of the tail is bent backwards and body
circulating filarial antigens like immunoradiometric
curves are few (Fig. 26.10).
assay and enzyme-linked immunosorbent assay (ELISA).
Brugia malayi: These measure about 230 μ × 6 μ in size. However, these tests are expensive and more labor-
Sheath stains dark pink in color. The nuclei are crowded intensive for routine use. Also, they cannot be applied in
in the body, are blurred in outline, and the tip of the tail field conditions.
240 Essentials of Clinical Pathology

Detection of Filarial DNA by Polymerase along Brahmaputra and Ganges, Bihar, Orissa, and
Chain Reaction Andhra Pradesh.
L. infantum is endemic in the Mediterranean basin and
Filarial DNA can be detected in circulating blood by
infection is zoonotic (i.e. transmitted to humans from
polymerase chain reaction. Although this test is highly
animals like dogs through sandfly vector).
sensitive and species-specific for diagnosis, it is laborious
L. chagasi occurs in Latin America with domestic dog
and expensive for routine use in developing countries.
as the primary reservoir of infection.
VISCERAL LEISHMANIASIS Typical manifestations of VL include fever, enlarge-
Leishmaniases are a group of parasitic diseases caused ment of spleen, enlargement of liver, severe cachexia,
by obligate intracellular protozoa of the genus Leishmania pancytopenia (anemia, leukopenia, and thrombo-
and transmitted by the bite of an infected female sandfly cytopenia), and hypergammaglobulinemia. Secondary
of the genus Phlebotomus (Africa, Asia, and Europe) or infections are frequent. The name “ Kala-azar” is derived
Lutzomyia (South and Central America). from the grayish coloration of skin that often develops
Leishmaniases are endemic in tropical and sub- during the course of disease. If untreated, death often
tropical regions of 88 countries on five continents. There follows.
are about 12 million cases of leishmaniases worldwide Post kala azar dermal leishmaniasis (PKDL) is a
and 1.5 to 2 million cases occur annually. Rising cutaneous form of leishmaniasis occurring after
prevalence of leishmaniases is attributed to massive rural resolution of visceral leishmaniasis. It is observed in India
to urban migration, deforestation with development of and East Africa. It manifests as hypopigmented and
newer dwelling sites, and newer irrigation projects. In raised erythematous lesions most prominently on the
addition, leishmania/human immunodeficiency virus face. The lesions contain amastigote forms of L. donovani.
coinfection is rapidly emerging as a new, severe disease. Since Leishmania are present in large numbers in
Leishmaniases occur in three different clinical forms: peripheral blood, they can be transmitted via sharing of
visceral, cutaneous, and mucocutaneous. Only visceral needles among intravenous drug abusers.
leishmaniasis is considered below. The typical clinical features of visceral leishmaniasis
In Asia, visceral leishmaniasis (VL) is also called as are not always seen and atypical presentations make
kala azar or black sickness. This is the most severe form of diagnosis difficult. Response to treatment is poor and
leishmaniases and is caused by Leishmania donovani relapses are common.
complex (which includes L. donovani, L. infantum, and L.
chagasi species). Majority of cases of VL occur in Life Cycle of Leishmania donovani
Bangladesh, northeastern Brazil, northeast India, Nepal,
and Sudan. Leishmania donovani (LD) exists in two stages: promasti-
In India, the causative organism is L. donovani and gote (flagellated form found in sandfly vector) and
infection is anthroponotic (i.e. transmitted from one amastigote (non-flagellated tissue-form found in
human to another through sandfly vector Phlebotomus). In mammalian host) (Fig. 26.11). Infection is transmitted to
India, leishmaniasis is prevalent in Assam and Bengal man when the infected female sandfly takes a blood meal

Fig. 26.11: Life cycle of Leishmania donovani


Diagnosis of Malaria and Other Parasites in Blood 241

and promastigote forms are inoculated. These promasti-


gotes are taken up by the macrophages of the reticulo-
endothelial system and are transformed into amastigote
forms. The amastigote forms multiply by binary fission
inside the macrophages, are released in circulation when
the host cell ruptures, and are again taken up by new
macrophages. In VL, whole of the reticuloendothelial
system is progressively infected (including bone marrow,
spleen, liver, lymph nodes, and blood monocytes).
When the female sandfly takes a blood meal, it ingests
intracellular and free amastigote forms present in blood.
These then develop into promastigote forms in the
midgut of the sandfly. After multiplication, promastigote Fig. 26.12: Leishmania donovani bodies in bone marrow
forms fill the lumen of the midgut and spread forwards (within a macrophage as well as lying free)
to the pharynx and buccal cavity. These are transmitted
to man when the infected female sandfly takes a blood
(Fig. 26.12). In buffy coat smears, they are seen inside
meal.
monocytes or less commonly inside neutrophils.
LABORATORY DIAGNOSIS OF VISCERAL Amastigote forms should be distinguished from yeast
LEISHMANIASIS forms of Histoplasma capsulatum (which show budding and
do not show kinetoplast). Splenic aspirate has the highest
Laboratory studies for diagnosis of VL include:
sensitivity for detection of tissue forms of Leishmania
• Examination of splenic aspirate, bone marrow
donovani. Since splenic aspiration is associated with a
aspirate, or buffy coat preparation of peripheral blood
risk of fatal hemorrhage, platelet count and prothrombin
for amastigote forms
time should be obtained before the procedure. Splenic
• Detection of anti-Leishmania antibodies
aspirate is contra-indicated if platelet count is <40,000/μl
• Intradermal skin test
and prothrombin time exceeds the control value by >5
• Culture of parasite
seconds.
• Animal inoculation studies
• DNA studies Tests for Detection of Anti-Leishmania Antibodies
Tests are available for detection of raised non-specific
Examination of Splenic Aspirate, Bone Marrow
immunoglobulins and specific anti-Leishmania anti-
Aspirate, or Buffy Coat Preparation of Peripheral
bodies.
Blood for Amastigote Forms
a. Tests that detect raised non-specific immuno-
The most commonly used method for demonstration of globulins: In VL, increased amounts of non-
the parasite is the examination of Giemsa- or Leishman- specific polyclonal IgG and IgM antibodies are
stained smears from relevant tissues. Usually, smear is produced. Tests, which detect these, are formol
prepared from splenic aspirate (sensitivity 95-98%), bone gel (aldehyde) test and antimony test. However,
marrow aspirate (sensitivity 60-85%), or buffy coat these tests are nonspecific and therefore unreliable
(sensitivity 67-99%). After staining, smears are examined for diagnosis of VL.
under oil-immersion lens of the light microscope for Formol gel (aldehyde) test can be used to support
amastigote forms (also called as LD bodies). Amastigote the diagnosis of VL. In this test, 1-2 drops of 40%
forms are small (2-4 μ in diameter), round to oval, and formalin solution are added to 1 ml of patient’s
have nucleus and a rod-shaped kinetoplast. Cytoplasm serum in a test tube. The mixture is allowed to
is pale blue while nucleus and kinetoplast stain pinkish stand for 2 hr. In a positive test, serum becomes
red. Nucleus is relatively large and kinetoplast lies at right milky white and gels (usually within 2-20 min)
angles to it. In aspirate smears, amastigote forms are seen (Fig. 26.13). This test becomes positive only when
in groups inside macrophages or lying free between cells the disease is of greater than 3 months duration.
242 Essentials of Clinical Pathology

positive in persons who have recovered from kala azar. It


is negative in active infection.

Parasite Culture
Cultures are usually made on Novy-McNeal Nicolle
(NNN) medium. After inoculation with 1-2 drops of
splenic or bone marrow aspirate, the culture is incubated
at 22 to 28°C, and examined weekly for promastigote
forms for upto 4 weeks.
In vitro culture of infected tissue is not required for
diagnosis in clinical practice. Parasite culture is necessary
if other methods of diagnosis are negative in the presence
Fig. 26.13: Formol gel test for visceral leishmaniasis
of strong clinical suspicion, and to obtain sufficient
quantity of antigen for direct agglutination test.

Animal Inoculation Studies


This test is also positive in other conditions with
hypergammaglobulinemia like multiple myeloma Laboratory animals (especially golden hamsters) are
and chronic liver disease. inoculated with infected tissue sample via intra-
b. Tests that detect specific anti-Leishmania anti- peritoneal, intrasplenic, or intradermal route. In positive
bodies in serum: Various tests have been developed cases, parasite can be demonstrated in cutaneous lesions,
for detection of anti-Leishmania antibodies like liver, or spleen.
complement fixation test, indirect immuno- The test becomes positive after several weeks or
fluorescence test, countercurrent immunoelectro- several months and therefore this test is not used for
phoresis, enzyme-linked immunosorbent assay, routine diagnosis.
direct agglutination test, and latex agglutination
test. DNA Diagnosis
In direct agglutination test (DAT), the antigen Molecular methods based on polymerase chain reaction
used is trypsin-treated promastigotes, which are have been described for diagnosis of VL. In this test, DNA
fixed in formalin and stained with Coomasie specific for Leishmania is identified. This test can be used
brilliant blue. Patient’s serum is incubated with for diagnosis, species identification, and in assessing
this antigen and agglutination is noted. Due to response to treatment. Currently this test is largely a
technical difficulties, the test is not widely research tool.
employed in field conditions.
A rapid and simple immunochromatographic BIBLIOGRAPHY
strip test has been developed for diagnosis of VL.
This test detects specific IgG anti-Leishmania 1. British Committee for Standards in Hematology. The
antibodies against rk39 antigen. This test has been laboratory diagnosis of malaria. Clin Lab Haem 1997;
19:165-70.
evaluated in field trials in India and is reported to
2. Cheesbrough M. District Laboratory Practice in Tropical
have high sensitivity and specificity. Countries. 1998, Cambridge University Press.
3. Chatterjee KD. Parasitology (Protozoology and
Skin Test Helminthology). 9th ed. Calcutta. Published by the
author, 1973.
The Montenegro (or Leishmanin) skin test is based on
4. Herwaldt BL. Leishmaniasis. Lancet 1999;354:1191-9.
delayed type of hypersensitivity reaction to Leishmaniasis. 5. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical
In this test, 0.5 ml of killed promastigotes are injected Hematology. (9th ed). London: Churchill Livingstone,
intradermally and the test is read after 72 hr. The test is 2001.
Diagnosis of Malaria and Other Parasites in Blood 243

6. Moody A. Rapid diagnostic tests for malaria parasites. 11. Weil GJ, Lammie PJ, and Weiss N. The ICT Filariasis
Clin Microbiol Reviews 2002;15:66-78. Test: A rapid-format antigen test for diagnosis of
7. Palmer CJ, et al. Evaluation of the OptiMal test for rapid bancroftian filariasis. Parasitology Today 1997;13:
diagnosis of P. vivax and P. falciparum malaria. J Clin 401-4.
microbial 1998;36:203-6. 12. WHO information Fact sheets: The Leishmaniases and
8. Sundar S, Rai M. Laboratory diagnosis of visceral Leishmania/HIV co-infections. Fact sheet no. 116.
Leishmaniasis. Clinical and Diagnostic Laboratory Revised May 2000. Geneva. World Health Organization.
13. World Health Organization. New perspectives in
Immunology 2002; 9:951-8.
malaria diagnosis. World Health Organization. 2000.
9. Warhurst DC, Williams JE. Laboratory diagnosis of
Geneva, Switzerland. WHO/MAL/2000.1091.
malaria. J Clin Pathol 1996;49:533-98. 14. World health Organization. Basic laboratory methods
10. Warrell DA, Gilles HM (Eds): Essential Malariology. (4th in medical parasitology. 1991. Geneva. World Health
Ed). 2002. London. Arnold. Organization.
27

Laboratory Tests in Anemia

Anemia is defined as a reduction in hemoglobin circulation causes palpitations and heart murmurs, and
concentration below the level, which is expected for in severe cases congestive cardiac failure can develop,
healthy persons of same age and sex, and in the same particularly in elderly. Anginal pain can result from
environment. Adequate oxygen cannot be delivered to myocardial hypoxia.
various organs and tissues due to low oxygen carrying Anemia is an objective sign of disease and needs
capacity of blood. further evaluation to determine the underlying cause and
The normal hemoglobin ranges, as proposed by appropriate treatment.
World Health Organization (WHO), are given at the end
of this chapter under “Reference Ranges”. CLASSIFICATION OF ANEMIAS
Anemia may occur without symptoms and may be
detected incidentally during medical examination. When There are two ways of classifying anemia:
severe enough, clinical features due to anemia result from • Etiological classification
hypoxia such as fatigue, weakness, dizziness, fainting, • Morphological classification
and mental confusion. Pallor of skin, mucous mem- 1. Etiological classification: Anemia can result from a
branes, and conjunctiva is present. Hyperdynamic variety of causes (Table 27.1).

Table 27.1: Etiological classification of anemia

I. Anemia due to decreased production of red blood cells


• Nutritional deficiencies: Iron deficiency anemia, megaloblastic anemia due to deficiency of folate or vitamin B12
• Anemia of chronic disease
• Sideroblastic anemia
• Aplastic anemia
• Anemia due to infiltration of bone marrow by malignant cells
• Anemia of chronic renal failure
II. Anemia due to increased destruction of red blood cells (Hemolytic anemia)
A. Hereditary:
• Defect in red cell membrane: Hereditary spherocytosis
• Defect in hemoglobin: Sickle cell disease, thalassemia, hemoglobin E disease
• Defect in red cell enzymes: Glucose-6-phosphate dehydrogenase deficiency
B. Acquired:
• Autoimmune hemolytic anemia
• Paroxysmal nocturnal hemoglobinuria
• Hemolytic transfusion reaction
• Hemolytic disease of newborn
• Mechanical hemolytic anemia
• Hypersplenism
• Malaria
III. Anemia due to acute blood loss: Hemorrhage due to trauma, massive gastrointestinal bleeding, or child delivery.
Laboratory Tests in Anemia 245

2. Morphological classification: This classification is Laboratory Features


based on red cell size and hemoglobin content (red
1. Low hemoglobin and low packed cell volume
cell indices) in a case of anemia. This classification is
2. Low mean cell volume, mean cell hemoglobin, and mean
given later in Table 27.16. cell hemoglobin concentration; red cell distribution
width is increased.
ANEMIAS DUE TO DECREASED
3. Blood smear shows microcytic, hypochromic red cells
PRODUCTION OF RED BLOOD CELLS
and pencil cells (Fig. 27.1).
Iron Deficiency Anemia 4. Serum ferritin is less than 15 μg/dl. The best
determinant of storage iron is serum ferritin. Serum
Iron deficiency is the most common cause of anemia ferritin >100 μg/dl rules out diagnosis of iron
worldwide, and is a public health problem in developing deficiency in most cases. Serum ferritin is a storage
countries. Pregnant women, women in reproductive age form of iron and is reduced only in iron deficiency
group, and children under 5 years of age are particularly anemia. However, it is also an acute phase reactant.
susceptible for nutritional deficiency. In men and If iron deficiency anemia is associated with infla-
postmenopausal women, blood loss (especially from the mmatory, neoplastic, or liver disease, its concen-
gastrointestinal tract) is the main cause. Common causes tration is raised; therefore, in these conditions,
are listed in Table 27.2. estimation of serum ferritin will not be helpful for
diagnosis of iron deficiency. In the presence of an
Table 27.2: Causes of iron deficiency anemia inflammatory disorder, assay for soluble transferrin
• Nutritional deficiency due to poor diet or increased receptor is preferable for diagnosis as it is not affected
requirements: infants and children (6 months-2 years), by inflammation.
women in reproductive age group, pregnancy 5. Serum iron, total iron binding capacity (TIBC), and
• Blood loss: transferrin saturation: Serum iron is a measure of iron
a. Gastrointestinal: Esophageal varices, peptic ulcer,
bound to transferrin (transport protein for iron).
carcinoma of stomach, hookworm infestation,
colorectal carcinoma, hemorrhoids
Serum iron level is affected by diurnal variation, iron-
b. Genitourinary tract: Menorrhagia, hematuria containing medications, hemolysis, etc. Total iron
• Malabsorption: Celiac disease binding capacity refers to the iron-binding sites of all
the circulating transferrin. Transferrin saturation is
the ratio of serum iron to transferrin. Typically, serum
Clinical Features iron is low, TIBC is increased, and transferrin
Apart from nonspecific clinical manifestations of anemia,
iron deficiency can cause koilonychia (flattened or spoon-
shaped nails), angular stomatitis, and glossitis.

Stages of Iron Deficiency


There are three stages of iron deficiency as shown in Box
27.1.

Box 27.1: Stages of iron deficiency

• Stage 1 (Iron depletion): Depletion of storage iron (low


serum ferritin), normal transport iron (serum iron, total
iron binding capacity), normal hemoglobin
• Stage 2 (Low transport iron): Depletion of storage iron,
low transport iron, normal hemoglobin
• Stage 3: (Low hemoglobin production): Depletion of
storage iron, low transport iron, low hemoglobin Fig. 27.1: Blood smear in iron deficiency anemia showing
microcytic hypochromic red cells and “pencil” cells
246 Essentials of Clinical Pathology

saturation is <10% in iron deficiency anemia. Free 8. Response to oral iron therapy: Increased reticulocyte
erythrocyte protoporphyrin is increased. count beginning around 3rd day and reaching
6. Increased soluble transferrin receptor (sTfR) in serum: This maximum on 5th-10th day after starting oral iron
assay is useful in cases with associated inflammation. therapy indicates optimal response. Hemoglobin rises
The transferrin receptor is a specific receptor for at the rate of 0.5-1.0 gm/dl/week.
transferrin-iron complex and is located on cell Iron deficiency anemia should be differentiated from
membranes. A soluble form also exists in circulation, other causes of microcytic hypochromic anemia such as
which is derived from proteolysis of cell membrane anemia of chronic disease, thalassemia, and sideroblastic
during erythrocyte maturation. Concentration of anemia (Table 27.3).
serum transferrin receptors correlates with the Diagnosis of iron deficiency anemia is straight
number of cellular receptors and varies with the rate forward. It is more important to determine the cause of
of erythropoiesis. In iron deficiency, its level increases iron deficiency, especially in adults since a serious
due to increased expression on cell membranes. underlying disorder (like carcinoma of gastrointestinal
However, increased levels are also found in any tract) may be present. To uncover the causative factor,
condition associated with increased erythropoietic apart from complete clinical examination, it may be
activity like hemolytic anemias and myeloprolife- necessary to perform stool examination for parasites and
rative disorders. for occult blood, urine examination for occult hematuria,
7. Bone marrow iron stain: This is the gold standard for radiologic and endoscopic workup of gastrointestinal
diagnosis of iron deficiency and shows lack of tract, and in females, pelvic ultrasonography.
stainable iron in the bone marrow. The test assesses
storage iron in bone marrow (confined mainly within Megaloblastic Anemia
macrophages; small amount in erythroblasts). This Megaloblastic anemia results from deficiency of either
test is expensive, and not required for routine folate or vitamin B12 (Table 27.4). Folate and vitamin B12
diagnosis. are essential for synthesis of deoxyribonucleic acid

Table 27.3: Differential diagnosis of microcytic


hypochromic anemia
Parameter Iron deficiency Anemia of β thalassemia Sideroblastic
anemia chronic disease minor anemia

1. Mean cell volume Low Normal or low Markedly low Low


2. Red cells on Microcytic Normocytic Marked Dimorphic
blood smear hypochromic normochromic; anisopoikilocytosis,
rarely microcytic basophilic stippling
hypochromic
3. Serum iron Low Low Normal Increased
4. Serum ferritin Low Normal or increased Normal Increased
5. TIBC Increased Low Normal Normal
6. Storage iron in marrow Absent Normal or increased Normal Normal
7. Hemoglobin electrophoresis Normal Normal Increased HbA 2 Normal
8. Iron in erythroblasts Absent Present Present Ringed sideroblasts
9. Serum soluble Increased Normal Normal or increased Normal or
transferrin receptor increased

TIBC: Total iron binding capacity


Laboratory Tests in Anemia 247

Table 27.4: Causes of megaloblastic anemia

Mechanism Folate deficiency Vitamin B12 deficiency

Inadequate intake Poor diet, chronic alcoholism Strict vegetarians


Inadequate absorption Malabsorption syndromes like Pernicious anemia*,
celiac disease, tropical sprue; gastrectomy, resection of ileum,
Crohn’s disease Crohn’s disease, fish tapeworm infestation
Increased demand Pregnancy, chronic
hemolysis, neoplasia –

*Autoimmune disorder characterized by gastric atrophy and loss of production of intrinsic factor in stomach that is
necessary for absorption of vitamin B12

(DNA); in case of deficiency, therefore, there is defective


synthesis of DNA in all the proliferating cells including
bone marrow cells. Vitamin B12 is also required for
neurological functions.
Anemia, mild jaundice (due to ineffective erythro-
poiesis or premature destruction of erythroid precursors
in bone marrow), and glossitis are common to both folate
and vitamin B12 deficiency. Neurologic features like
symmetric paresthesiae especially in lower limbs,
reduced vibration sense, ataxia, loss of memory, and
personality change occur in vitamin B12 deficiency but
not in folate deficiency. Untreated cases may develop
subacute combined degeneration of the spinal cord.
Neurological damage due to vitamin B12 deficiency can
occur in the absence of anemia.

Laboratory Features Fig. 27.2: Blood smear in megaloblastic anemia showing


oval macrocytes and a hypersegmented neutrophil
• Macrocytic anemia (mean cell volume >100 fl in
adults). Elevation of mean cell volume is an early sign
and precedes the onset of anemia. Pancytopenia is
common.
• Blood smear shows oval macrocytosis, basophilic
stippling, Howell-Jolly bodies, and hyperseg-
mentation of neutrophils (>5% of neutrophils
showing 5 or more lobes) (Fig. 27.2).
• Reticulocyte count is normal or low.
• Bone marrow shows megaloblasts, erythroid hyper-
plasia, and giant metamyelocytes and bands
(Fig. 27.3).
• Vitamin assays: See Table 27.5.

Identification of cause It is necessary to distinguish


between folate and vitamin B12 deficiency and to Fig. 27.3: Bone marrow smear showing megaloblasts and
determine the cause for appropriate treatment a giant metamyelocyte
248 Essentials of Clinical Pathology

Fig. 27.4 Evaluation of suspected megaloblastic anemia. (*: Measurement of serum methylmalonic acid is said to be
more sensitive than measurement of vitamin B12 and an earlier marker for detection of vitamin B12 deficiency)

Table 27.5: Differences between folate and vitamin B12 deficiency

Parameter Folate deficiency Vitamin B12 deficiency

1. Usual mechanism of deficiency Inadequate intake Inadequate absorption


2. Neurologic features Absent May be present
3. Serum vitamin B12 Normal Low
4. Serum folate Low Normal or increased
5. Red cell folate Low Low
6. Serum homocysteine Increased Increased
7. Serum methylmalonic acid Normal Increased
8. Therapeutic trial Optimal response to folate Optimal response to vitamin B12

(Table 27.5, and Fig. 27.4). Treatment only with folate in Anemia of Chronic Disease
vitamin B12 deficiency can worsen the neurological Anemia of chronic disease is the most common form of
abnormalities. anemia amongst hospitalized patients. The three disease
In vitamin B 12 deficiency, test for vitamin B 12 categories associated with anemia of chronic disease are
absorption (Schilling test) can be carried out. In this test, chronic infection, inflammation, and malignancy
(Table 27.6). There is a block in release of storage iron
urinary excretion of oral dose of radio-labeled vitamin
from macrophages for erythropoiesis that is mediated
B 12 is compared with the excretion of oral dose of by inflammatory cytokines.
radiolabeled vitamin B12 bound to intrinsic factor. In Anemia is mild (9-10 gm/dl) and non-progressive.
pernicious anemia, deficient absorption is corrected with Clinical features reflect underlying disease. The main
addition of intrinsic factor. differential diagnosis is iron deficiency anemia.
Laboratory Tests in Anemia 249

Table 27.6: Diseases associated with anemia of chronic disease


1. Chronic infections: Tuberculosis, urinary tract infection, bronchiectasis, osteomyelitis, subacute bacterial
endocarditis
2. Chronic inflammation: Rheumatoid arthritis, systemic lupus erythematosus.
3. Malignancy

Laboratory Features Sideroblastic anemia may be hereditary (X-linked) or


• Normocytic normochromic anemia (70% cases) or acquired. Most cases are acquired and causes include:
microcytic hypochromic anemia (30% cases). (i) drugs: isoniazid, chloramphenicol, cytotoxic drugs,
• Decreased serum iron, decreased total iron binding (ii) alcoholism, (iii) lead poisoning, (iv) myelodysplastic
capacity, and normal or raised serum ferritin. Serum syndrome, and (v) acute myeloid leukemia.
transferrin receptor level is normal. Aplastic Anemia
• Increased marrow storage iron
• Erythrocyte sedimentation rate is increased out of Aplastic anemia is characterized by pancytopenia in
proportion to the degree of anemia. peripheral blood (reduction of red cells, leukocytes, and
platelets in peripheral blood) and decreased cellularity
Sideroblastic Anemia in bone marrow. Aplastic anemia can occur at any age
In sideroblastic anemia, heme synthesis is deficient. There and clinical presentation is related to pancytopenia
is a mitochondrial defect that leads to the failure of (anemia, risk of infections, and bleeding manifestations).
incorporation of iron into heme. Iron accumulates in Organomegaly is typically absent.
mitochondria that surround the nucleus of erythroblasts Causes of aplastic anemia are listed in Table 27.7.
forming ringed sideroblasts. Sideroblastic anemia is
Investigations in a Suspected Case of
characterized by:
Aplastic Anemia
• Dimorphic anemia (i.e. blood smear shows dual
population of cells: normocytic normochromic, and • Tests for confirmation of aplastic anemia: Complete blood
microcytic hypochromic) (Fig. 27.5). count (to demonstrate pancytopenia, low hemo-
• Ringed sideroblasts in bone marrow (see Fig. 27.5). globin, and low reticulocyte count) and bone marrow
examination (to demonstrate depletion of hema-
topoietic precursors and hypocellularity).

Table 27.7: Causes of aplastic anemia

Acquired

Idiopathic
Drugs:
Idiosyncratic: antibacterials (chloramphenicol,
sulfonamides), nonsteroidal anti-inflammatory drugs
(phenylbutazone, indomethacin, piroxicam, diclofenac),
antithyroid drugs, furosemide, phenothiazines, allopurinol,
oral antidiabetics, Dose-related: Cytotoxic drugs
Chemicals (e.g. benzene) or radiation
Infections: Hepatitis; Epstain Barr virus, Mycobacteria
Paroxysmal nocturnal hemoglobinuria
Systemic lupus erythematosus
Graft vs. host disease

Fig. 27.5: Blood smear (on left) in sideroblastic anemia showing Inherited
dimorphic red cells, basophilic stippling, and a polychromatic
Fanconi’s anemia, Dyskeratosis congenita
red cell. Bone marrow smear stained with iron stain (on right)
shows ringed sideroblasts
250 Essentials of Clinical Pathology

• Tests for diagnosis of underlying cause: Viral studies polypeptide chain with abnormal structure (Table 27.8).
(hepatitis A antibody, hepatitis B surface antigen, The inherited disorders of hemoglobin have achieved a
hepatitis C antibody), Ham’s test and/or flow high frequency due to the selective advantage afforded
cytometry for lack of CD55 and CD59 (for paroxysmal to the heterozygotes of these genetic variations against
nocturnal hemoglobinuria), detailed drug history, infection by Plasmodium falciparum.
antinuclear antibody test (for systemic lupus
erythematosus), and cytogenetic analysis (for Fanconi Thalassemias
anemia). Etiology remains unknown in 50% cases of
The thalassemias are inherited disorders characterized
aplastic anemia.
by reduced or absent synthesis of α or β globin
Severity of Aplastic Anemia polypeptide chains.

1. Severe aplastic anemia: Aplastic anemia is considered to Classification


be severe when following features are present (Camitta There are two main forms of thalassemias:
et al, 1976): • α thalassemias (deficient synthesis of α globin chains).
• Bone marrow cellularity is either < 25% of normal or • β thalassemias (deficient synthesis of β globin chains).
is 25-50% of normal with <30% of hematopoietic cells,
AND Distribution
• Two out of three of the following:
• α thalassemias: Southeast Asia, Eastern Mediterranean
– Reticulocytes <1%
region, Middle East.
– Neutrophils <500/cmm
• β thalassemias: Mediterranean region, Africa, Middle
– Platelets <20000/cmm
East, India, Pakistan, Southeast Asia.
2. Very severe aplastic anemia (Bacigalupo et al, 1988): In India, β thalassemia is more common in commu-
Criteria are as above, except neutrophil count is < 200/ nities like Punjabis, Sindhis, Bengalis, Gujaratis,
cmm. Bhanushalis, and Jains.
3. Non-severe aplastic anemia: This is present when criteria
β thalassemias: Normally, there are two β globin genes,
do not meet requirements for severe or very severe
one on each member of chromosome 11 (genotype β/β).
aplastic anemia.
β thalassemias result from a single base substitution
(point mutation) in β globin gene. More than 150
HEREDITARY DISORDERS OF
mutations of β globin gene have been reported to cause
HEMOGLOBIN
β thalassemia. There are two main forms of β
Hereditary disorders of hemoglobin result either from thalassemias—β0 and β+. β0 thalassemia is characterized
(i) reduced synthesis of one globin polypeptide chain, by complete absence of β chain synthesis, while in β+
leading to chain imbalance, or (ii) synthesis of a thalassemia β chain synthesis is reduced.

Table 27.8: Classification of hereditary disorders of hemoglobin


Disorder Distribution Basic defect Examples

1. Hemoglobinopathies Varied; In India, HbS, Structural alteration of a HbS, HbD, HbC


HbD, and HbE are common globin polypeptide chain
2. Thalassemias Mediterranean region, Reduced synthesis of one α-thalassemia,
Africa, Middle east, globin polypeptide chain β-thalassemia
India, Pakistan,
Southeast Asia
Laboratory Tests in Anemia 251

There are three clinical forms of β thalassemias:


thalassemia major, thalassemia intermedia, and
thalassemia minor.
1. β thalassemia major (Homozygous β thalassemia, Cooley’s
anemia): β thalassemia major results when both the β
globin genes are defective, e. g. genotype β°/β° or
β° /β+ . It is characterized by severe anemia, and
requires regular blood transfusion therapy for
survival.
Clinical features of β thalassemia major are severe
hemolytic anemia that develops around 6 months of age,
jaundice and progressive splenomegaly, severe growth
retardation and delayed development, skeletal defor-
mities (frontal bossing, malar prominence), susceptibility
to infections and to folate deficiency, and transfusion- Fig. 27.6: Blood smear in β thalassemia major showing marked
dependence for survival. anisopoikilocytosis, microcytosis, and hypochromia
In untreated patients, death usually occurs before 5
years of age. In individuals who receive regular blood • Blood smear shows less severe abnormalities than
transfusion therapy, iron overload gradually develops thalassemia major
during adolescence. Unless iron chelation therapy is • Reticulocytosis (5-10%)
given, such individuals will die prematurely from • Varible elevation of hemoglobin F on electrophoresis
damage to organs like heart, pancreas, and liver. 3. β thalassemia minor (thalassemia trait, asymptomatic
thalassemia): Usually individuals having one normal
Characteristic laboratory features are:
and one abnormal β globin gene have β thalassemia
• Severe anemia (hemoglobin < 7.0 gm/dl).
minor (e.g.β/β+ or β/β°). In this heterozygous carrier
• Blood smear shows marked variation in size and
state, anemia is either mild or absent. This condition
shape of red cells, red cell fragments, hypochromia,
is often detected during the course of routine
target cells, basophilic stippling, and nucleated red
hematological investigations. β thalassemia trait
cells (Fig. 27.6).
should be distinguished from iron deficiency anemia
• Reticulocytosis (5-15%).
as iron therapy is not required in the former. Serum
• Low mean cell volume, mean cell hemoglobin, and
iron and serum ferritin are low in iron deficiency
mean cell hemoglobin concentration.
anemia, while normal in β thalassemia minor.
• Markedly elevated hemoglobin F on electrophoresis
(Fig. 27.28, later). Laboratory features are:
2. β thalassemia intermedia: This results from homozygous • Hemoglobin slightly low or normal (> 10 gm/dl)
inheritance of mild β+ thalassemia (e.g. genotype β+/ • Low mean cell volume and mean cell hemoglobin;
β+). It is characterized by moderate degree of anemia normal mean cell hemoglobin concentration
which does not require regular blood transfusion • Blood smear shows microcytic hypochromic red cells,
target cells, and basophilic stippling (Fig. 27.7)
therapy. Worsening of anemia occurs during
• Normal serum ferritin
infections and pregnancy.
• Hemoglobin electrophoresis shows elevated hemo-
Thalassemia intermedia presents at a later age (2-5
globin A2 (> 3.5%); this is a diagnostic test for β
years) than thalassemia major. These patients have
thalassemia minor.
chronic hemolytic anemia, splenomegaly, and skeletal
In Indian communities with a high prevalence of
changes.
β thalassemia gene, spouses of persons with β thala-
Laboratory features are: ssemia trait should be screened for β thalassemia trait
• Moderate degree of anemia (hemoglobin 7-10 and hemoglobin E trait to avoid birth of a child with
gm/dl). homozygous β thalassemia.
252 Essentials of Clinical Pathology

There are three clinical forms of α thalassemia: Hb


Bart’s hydrops fetalis syndrome (--/--), Hb H disease (--
/-α), and α thalassemia carrier state (-α/αα, -α/-α, or --
/αα) (Fig. 27.8).
Hb Bart’s hydrops fetalis syndrome is the most severe
form and infants are either stillborn prematurely or die
soon after birth. They have severe anemia, generalized
edema, and hepatosplenomegaly. Mothers of such
infants usually have toxemia of pregnancy. Blood smear

Fig. 27.7: Blood smear in β thalassemia minor showing


microcytes with hypochromia, target cells, polychromatic cells,
and basophilic stippling

β thalassemia major and minor are compared in Table


27.9.
α thalassemias: Normally, there are four α globin genes,
two on each member of chromosome 16 (genotype αα/
αα). α thalassemias usually results from gene deletions.
Since α chains are present in both fetal and adult
hemoglobin, α thalassemia manifests in both fetal and Fig. 27.8: Genotypes in α thalassemias. Normal genotype is
represented as αα/αα. The effective genotypes of various
adult lives. In α° thalassemia, both α globin genes on
thalassemia syndromes are represented as: Hb Bart’s hydrops
one chromosome are lost, while in α+ thalassemia, one α fetalis syndrome: --/--; Hb H disease: α-/--; and α thalassemia
globin gene of the pair is lost. carrier state: αα/-- or αα/α-

Table 27.9: Comparison of two main forms of β thalassemias

Parameter β thalassemia major β thalassemia minor

1. Genotype β0/β+, β0/β 0 β/β+, β/β0


2. Blood smear Marked anisopoikilocytosis, Microcytosis, hypochromia, target
microcytosis, hypochromia, cells, basophilic stippling
target cells, basophilic
stippling, nucleated red cells
3. Hemoglobin electrophoresis β0/β0: HbA: 0, HbF: 95-98%, HbA: 90-95%, HbF: 0.5-4%,
HbA2: 2-5%; β0/β+: HbA: 10-30%, HbA2: >3.5%
HbF: 70-90%, HbA2: 2-5%

Note: β0 indicates no production of β globin chain; β+ indicates diminished but some production of β globin chain;
β indicates normal β chain production
Laboratory Tests in Anemia 253

shows marked variation in size and shape of red cells, Mode of inheritance is autosomal recessive. If both the
microcytosis, hypochromia, and nucleated red cells. parents have sickle cell trait, sickle cell anemia will develop
Hemoglobin electrophoresis reveals predominance of Hb in the offspring (1:4 chance). Clinical features of sickle cell
Bart’s and absence of Hb A. anemia are highly variable, and range from infrequent
Patients with HbH disease have moderate anemia and mild to frequent and severe. Manifestations include
(hemoglobin 7-10 gm/dl), jaundice, and hepato- chronic hemolytic anemia, vaso-occlusive crises (acute
splenomegaly. Typical laboratory features are microcytic episodes of severe pain in chest, abdomen, back, or
hypochromic red cells, Hb H inclusions in red cells, and extremities), aplastic crisis (sudden aggravation of anemia
variable amount of HbH on electrophoresis. due to infection by parvovirus), hemolytic crisis
α thalassemia carriers are asymptomatic. Microcytic (exacerbation of hemolysis due to infection or associated
hypochromic red cells may or may not be present. glucose-6-phosphate dehydrogenase deficiency), splenic
Hemoglobin electrophoresis is normal. Determining sequestration crisis (sudden enlargement of spleen due to
ethnic origin, family studies, globin chain analysis, or pooling of blood leading to circulatory failure), risk of
genetic analysis may be needed for definitive diagnosis. infections (especially Streptococcus pneumonia, Hemophilus
influenzae, Neisseria meningitidis, Escherichia coli) and
Sickle Cell Disorders retarded growth and development.
In sickle cell anemia, blood smear shows sickled red
Sickle cell disorders are characterized by the presence of cells and target cells (Fig. 27.9), solubility test is positive,
hemoglobin S (HbS) in red cells due to a point mutation and hemoglobin electrophoresis shows mostly HbS with
A→T in the 6th codon of β globin gene, which results in no HbA. In sickle cell trait, blood smear is normal or
substitution of valine for glutamic acid at position 6 of β shows target cells, solubility test is positive, and
polypeptide chain (β6glu→val). Upon deoxygenation, HbS hemoglobin electrophoresis shows HbA (60%) and HbS
polymerizes in red cells leading to the formation of sickle- (40%).
shaped red cells; such red cells are rigid, occlude the Although blood smear shows sickle cells in sickle cell
microvasculature, and cause painful vaso-occlusive anemia, and both slide test and solubility test are positive
crises. Sickle cells adhere to vascular endothelium due in sickle cell disorders, hemoglobin electrophoresis is
to increased expression of adhesion molecules. Deformed
red cells are removed from the circulation and destroyed
prematurely, leading to chronic hemolytic anemia.
Disease manifests in individuals with homozygous sickle
cell disease; heterozygotes are usually asymptomatic.
Sickling disorders (Table 27.10) are prevalent in
Africa, Middle East, Mediterranean region, and central
and southern parts of India. It is thought that the disease
has achieved high frequency due to evolutionary
selection for resistance against falciparum malaria.

Table 27.10: Sickle cell disorders

Disorder Genotype

1. Sickle cell anemia βS/βS


2. Sickle cell trait βS/β
3. Double heterozygous states
• Hemoglobin S-C disease βS/βC
• Hemoglobin S-D disease βS/βD
• Sickle cell-β thalassemia βS/β0
Fig. 27.9: Blood smear in sickle cell anemia.
254 Essentials of Clinical Pathology

Table 27.11: Hemoglobin electrophoresis patterns in sickle cell disorders

Disorder Hemoglobin Parents’ phenotype


electrophoresis

1. Sickle cell anemia SF Both parents: AS


2. Sickle cell trait AS One parent: AA; Other parent: AS
3. Sickle cell-β0 thalassemia SF; A2 increased One parent: AS; Other parent: β thalassemia minor
4. Sickle cell-β+ thalassemia SAF; A2 increased One parent: AS; Other parent: β thalassemia minor

essential for diagnosis (Fig. 27.28, later). This is to


differentiate between sickle cell trait and sickle cell
anemia, and for detection of double heterozygous states
like sickle cell-β thalassemia or sickle cell-HbD disease
(Table 27.11).

Hemoglobin D
In India, hemoglobin D is observed in Punjab. Hetero-
zygotes (A/D) are asymptomatic, while homozygotes
(D/D) have mild anemia. Blood smear shows hypo-
chromia, microcytosis, and target cells. On hemoglobin
electrophoresis at alkaline pH, hemoglobin D co-migrates
with HbS. Slide test for sickling, however, is negative in
Hb D disease.

HEREDITARY SPHEROCYTOSIS
Fig. 27.10: Blood smear in hereditary spherocytosis showing
This is the most common type of hereditary hemolytic spherocytes, a polychromatic cell, and a nucleated red cell
anemia in North Europeans. Hereditary spherocytosis
(HS) is characterized by an inherited defect in the red
cell cytoskeleton leading to the formation of spherocytes. Anemia, reticulocytosis, and microspherocytes on
Normally, the lipid bilayer is anchored to the underlying blood smear are the usual findings (Fig. 27.10). Micro-
cytoskeleton by interactions of (i) spectrin, protein 4.1, spherocytes are small and dense red cells lacking central
actin, and glycophorin C, and (ii) spectrin, ankyrin, and area of pallor. (They are also observed in autoimmune
band 3. In most cases of HS, there is a deficiency of hemolytic anemia, ABO hemolytic disease of newborn,
ankyrin so that membrane becomes unstable with burns, microangiopathic hemolytic anemia, and
fragmentation of part of membrane, loss of surface area, hemolytic transfusion reaction).
and spherocyte formation. Spherocytic red cells are rigid, The usual screening test for HS is osmotic fragility
less deformable than normal red cells, and are destroyed (OF) test (Figs 27.31 and 27.32, later). This test assesses
prematurely in spleen. Mode of transmission is usually the ability of the red cells to withstand osmotic stress
autosomal dominant. when they are suspended in decreasing concentrations
Usual clinical manifestations are presentation in of hypotonic saline solutions. Spherocytes have reduced
childhood with mild to moderate anemia, intermittent surface area to volume ratio and are osmotically fragile.
jaundice, and splenomegaly. Pigment gallstones are Incubated variant of OF test is more sensitive. Auto-
frequent. Similar history is obtained in a close relative. hemolysis test shows markedly increased hemolysis that is
Clinical presentation, however, is markedly variable. partially corrected by addition of glucose (Fig. 27.33, later).
Laboratory Tests in Anemia 255

Direct antiglobulin test is carried out to exclude immune


hemolysis.

GLUCOSE-6-PHOSPHATE
DEHYDROGENASE DEFICIENCY

Glucose-6-phosphate dehydrogenase (G6PD) deficiency


is an X-linked disorder characterized by reduced activity
of G6PD enzyme in red cells and occurrence of hemolysis
on exposure to oxidant stress. In India, G6PD deficiency
is especially common in Parsees and Vataliya Prajapatis.
Numerous (>400) biochemical variants of G6PD enzyme
have been reported due to point mutations or deletions
in the gene. In India, G6PD Mediterranean, G6PD Kerala-
Kalyan, and G6PD Orissa variants are more common.
G6PD deficiency leads to inability of red cells to
remove toxic hydrogen peroxide (H2O2), an oxidative Fig. 27.11: Blood smear during acute hemolysis in glucose
metabolite. Accumulated H2 O2 leads to oxidation of 6 phosphate dehydrogenase deficiency showing a bite cell
hemoglobin with subsequent denaturation and precipi- and hemi-ghost cells
tation of globin chains. Precipitated globin form
inclusions attached to red cell membrane (Heinz bodies).
(due to intravascular hemolysis). Heinz bodies can be
Red cells containing Heinz bodies are destroyed in spleen
demonstrated by methyl violet staining. The screening
(extravascular hemolysis). Oxidant damage also causes tests used for G6PD deficiency are fluorescent spot test,
peroxidation of membrane lipids of red cells leading to methemoglobin reduction test, and dye decolorization
intravascular hemolysis. test.
G6PD deficiency is usually asymptomatic. It can
cause (i) neonatal jaundice, (ii) acute hemolytic anemia IMMUNE HEMOLYTIC ANEMIAS
on exposure to oxidant stress (Table 27.12), or (iii) chronic
Immunological destruction of red cells occurs when
hemolytic anemia.
antibody and/or complement bind to red cell membrane.
Examination of blood smear during hemolytic
Hemolysis may be extra- or intravascular. Classification
episode shows polychromasia, fragmented red cells,
of immune hemolytic anemias is shown in Table 27.13.
spherocytes, bite cells (red cells having bitten out
Warm antibody type autoimmune hemolytic anemia
margins) and half-ghost cells (one half of the red cell is
occurs mainly in persons over 50 years of age. Mild
empty and the other half is filled with hemoglobin)
jaundice and splenomegaly are common. Red cells coated
(Fig. 27.11). Biochemical abnormalities include increased
with IgG are recognized by Fc receptors on macrophages
serum bilirubin, hemoglobinemia, and hemoglobinuria
and phagocytosed in spleen. In many cases, phagocytosis
is incomplete with formation of spherocytes (Fig. 27.12).
Red cells coated with IgG are detected by direct
Table 27.12: Causes of hemolysis in glucose-6- antiglobulin test (Fig. 27.34, later).
phosphate dehydrogenase deficiency
Cold agglutinin disease is characterized by acro-
1. Bacterial and viral infections cyanosis (cyanosis of fingers, toes, nose, and ears) due to
2. Drugs: the presence of cold agglutinins that cause agglutination
• Antimalarials: Primaquine, pamaquine of erythrocytes on exposure to cold. The antibody is IgM,
• Antibacterials: Sulfonamides, nalidixic acid, cold-reactive, and after binding to red cells activates
nitrofurantoin, dapsone complement. The disease usually occurs in older
• Antipyretics and analgesics
individuals. Blood smear shows autoagglutination of red
3. Chemicals: Naphthalene balls cells (Fig. 27.13); preparation of blood smear after
256 Essentials of Clinical Pathology

Table 27.13: Classification of immune hemolytic anemias


1. Autoimmune hemolytic anemia
• Warm antibody type
– Primary or idiopathic
– Secondary: infections, autoimmune disorders, lymphoma, chronic lymphocytic leukemia
• Cold antibody type
– Cold agglutinin disease
– Paroxysmal cold hemoglobinuria
2. Alloimmune hemolytic anemia
• Hemolytic disease of newborn
• Hemolytic transfusion reaction
3. Drug-induced hemolytic anemia

Fig. 27.12: Blood smear in warm antibody type autoimmune Fig. 27.13: Blood smear in cold agglutinin disease
hemolytic anemia showing spherocytes, polychromasia, and showing large clusters of red cells (autoagglutination)
late normoblast

warming of blood sample causes disappearance of case of anti-D, mother is Rh D-negative and father is Rh
autoagglutination. Direct antiglobulin test is positive due D-positive. Rh HDN develops during second or
to the presence of complement on red cells. subsequent pregnancies if the fetus is Rh D-positive.
Clinical presentation is variable. Rh HDN may
HEMOLYTIC DISEASE OF NEWBORN manifest with (i) mild anemia and jaundice, (ii) icterus
Hemolytic disease of newborn (HDN) is characterized gravis neonatorum with kernicterus, or (iii) hydrops
by destruction of red cells of the fetus or neonate due to fetalis with intrauterine fetal death.
antibodies produced by the mother. Allo-antibodies The usual clinical presentation of ABO HDN is mild
develop in the mother against foreign red cell antigens anemia and jaundice.
of the fetus inherited from the father. Leakage of fetal Antenatal investigations consist of:
red cells during pregnancy or delivery into the maternal 1. Maternal: These are ABO and Rh grouping, antibody
circulation stimulates formation of antibodies. Passage screening, antibody identification, and antibody
of IgG maternal antibodies across placenta into the fetal titration: If antibody is clinically significant, titer is
circulation causes hemolysis of fetal red cells. determined. Titer >1:32 or a rising titer showing
The two main red cell antigens responsible for HDN increase of 2 dilutions or more is significant and
are Rh and ABO. In Rh HDN, antibodies develop against amniocentesis should be performed to determine
anti-D and less commonly against anti-C or anti-E. In severity of disease (Fig. 27.14).
Laboratory Tests in Anemia 257

Fig. 27.14: Approach to the management of hemolytic disease of newborn

2. Fetal: This is amniocentesis or cordocentesis for


assessment of severity of hemolysis by determining
bilirubin level. Liley’s graph is used for predicting
severity of HDN and management of the fetus. Liley’s
graph plots degree of absorption at 450 nm versus
gestational age in weeks on a semilogarithmic graph
paper.
After delivery, Kleihauer-Betke acid elution test
(Fig. 27.15) is used to identify number of fetal red cells in
maternal circulation, and calculate the number of vials
of Rh immune globulin to be infused to prevent maternal
immunization (Fig. 27.16).

Investigations in newborn consist of:


1. Blood grouping
2. Blood smear (Fig. 27.17)
3. Direct antiglobulin test (DAT) on cord red cells:
Fig. 27.15: Acid elution or Kleihauer-Betke test. Maternal blood
Positive DAT (Fig. 27.34, later) can result from coating sample is withdrawn within 2 hours of birth, a blood smear
of red cells of the newborn with immune anti-A or is prepared and flooded with acid. Hemoglobin in adult red
anti-B from group O mother, or immune anti-D from cells (HbA) is washed out by the acid solution, while hemoglobin
Rh-negative mother. in fetal red cells (HbF) is not. After counterstaining with safranin,
red cells containing HbA appear pale, while cells containing
4. Estimation of bilirubin and hemoglobin
HbF appear dark. Acid elution test is performed to assess
Differences between Rh and ABO HDN are presented amount of fetomaternal hemorrhage and to calculate the dose
in Table 27.14. of Rh immune globulin
258 Essentials of Clinical Pathology

Fig. 27.16: Use of Kleihauer-Betke test to determine the dose of anti-D following delivery by a Rh D-negative woman
of a Rh D-positive baby. The maternal sample should be taken within 2 hours of delivery or any other sensitizing event

PAROXYSMAL NOCTURNAL
HEMOGLOBINURIA
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare,
acquired clonal stem cell disorder in which red cells are
abnormally sensitive to the hemolytic action of the
complement. PNH red cells are deficient in several cell
membrane proteins including CD 55 (decay accelerating
factor) and CD 59 (membrane inhibitor of reactive lysis or
MIRL). Normally, small amount of complement is being
continuously generated via alternate complement
pathway. CD 55 inactivates C3 convertase while CD 59
inhibits the formation of membrane attack complex and
protects the red cells against complement-mediated attack.
Fig. 27.17: Blood smear in Rh hemolytic disease of
In a typical case of PNH, red cell destruction occurs newborn showing erythroblasts and polychromatic cells
at night so that hemoglobinuria (passage of reddish-
brown urine) is noticed after getting up in the morning.
However, presentation is often variable and may be in • Demonstration of increased sensitivity of red cells to
the form of pancytopenia, aplastic anemia, chronic complement: Sucrose hemolysis test, Ham’s test.
intravascular hemolysis, or recurrent venous thromboses. • Flow cytometric analysis of blood cells for lack of
Some cases of PNH evolve to acute myeloid leukemia. CD55 or CD59.
Laboratory diagnosis of PNH is based on:
• Evidence of intravascular hemolysis: Hemoglo- MICROANGIOPATHIC HEMOLYTIC ANEMIA
binuria, hemosiderinuria, methemalbuminemia, and This is a form of mechanical hemolytic anemia resulting
increased indirect serum bilirubin. from intravascular fragmentation and lysis of red cells
Laboratory Tests in Anemia 259

Table 27.14: Comparison of Rh and ABO hemolytic disease of newborn


Parameter Rh HDN ABO HDN

1. Blood group of mother D-negative O


2. Blood group of fetus D-positive A or B
3. Pregnancy usually affected Second or subsequent First
4. Hydrops fetalis Common Uncommon
5. Stillbirth Common Uncommon
6. Severe anemia Usual Rare
7. Blood smear Erythroblastosis Spherocytes
8. Direct antiglobulin test Strongly positive Weakly positive or negative
9. Prevention Rh immune globulin None

diagnosis of anemia are estimation of hemoglobin and


packed cell volume.

Estimation of Hemoglobin
Cyanmethemoglobin method is the method of choice.
Recently introduced WHO hemoglobin color scale is a
simple and inexpensive method and well suited for
under-resourced laboratories (see Chapter 18: Estimation
of Hemoglobin).
Depending on hemoglobin concentration, anemia is
graded as mild (hemoglobin less than lower limit of
normal but above 10.0 gm/dl), moderate (hemoglobin
7-10 gm/dl), and severe (hemoglobin < 7 gm/dl).
Measured hemoglobin level depends on amount of
hemoglobin in red blood cells and blood volume.
Fig. 27.18: Blood smear in microangiopathic hemolytic Pseudoanemia or spurious anemia results from relative
anemia showing many fragmented red cells increase in plasma volume in pregnancy, splenomegaly,
congestive cardiac failure, and paraproteinemias.
Therefore, hemoglobin level should be interpreted in the
due to vascular endothelial abnormalities. Common
light of clinical features.
causes include thrombotic thrombocytopenic purpura,
hemolytic uremic syndrome, disseminated intravascular Estimation of Packed Cell Volume (PCV)
coagulation, eclampsia, disseminated malignancy, and
PCV is the volume of packed red cells obtained after
generalized vasculitis. Blood film shows numerous
centrifugation of a sample of anticoagulated blood in a
fragmented red cells (schistocytes) (Fig. 27.18).
Wintrobe tube or a capillary tube (see Chapter 19 : Packed
APPROACH TO DIAGNOSIS OF ANEMIA Cell Volume). Hematocrit is the equivalent measurement
derived on automated blood cell analyzer from the red
Evaluation of a case of anemia consists of: cell indices. Although not entirely synonymous, the terms
• Establishing the presence and severity of anemia, and PCV and hematocrit are used interchangeably in clinical
• Determining the cause of anemia. practice.
PCV is about three times the value of hemoglobin
Establishing the Presence and concentration and thus can be used to cross-check the
Severity of Anemia
hemoglobin value.
Clinical features of anemia are non-specific and their Sometimes, additional information can be gained
assessment is often subjective. Laboratory methods for from the observation of color of plasma and thickness of
260 Essentials of Clinical Pathology

buffy coat layer. Normal plasma is straw-colored; it is


colorless in iron deficiency anemia, pink in hemo-
globinemia, and yellow in hemolysis. A thick buffy coat
indicates leukocytosis and/or thrombocytosis.

Determining the Cause of Anemia

Determining the cause of anemia depends on evaluation


of clinical and laboratory studies (Fig. 27.19).

Evaluation of Clinical Features in Anemia

Clinical features in a case of anemia depend on (i) severity


of anemia, (ii) cause of anemia, and (iii) rapidity of
development of anemia. Anemia may be acute or chronic.
Acute anemia may be due to either acute blood loss or
acute hemolysis, and symptoms are often due to loss of
circulatory volume. Chronic anemia is tolerated well due Fig. 27.19: Approach to diagnosis of anemia
to compensatory mechanisms. Chronic anemia may be
symptomatic or may be detected incidentally during the
Basic Laboratory Studies in Anemia
course of investigations for some other disease. Clinical
features, which are related to the cause of anemia, are Peripheral blood smear: In majority of cases, if correlated
shown in Table 27.15. with clinical features, blood smear findings suggest the

Table 27.15: Clinical features and the cause of anemia

Clinical feature Probable cause of anemia

1. Pregnancy, females of reproductive age group, Nutritional deficiency


growing children
2. Chronic blood loss Iron deficiency
3. Chronic alcoholism Folate deficiency, gastrointestinal blood loss,
sideroblastic anemia
4. Ingestion of drugs:
• Prolonged intake of aspirin Iron deficiency from gastric bleeding
• Cytotoxic drugs Aplastic anemia
• Alpha methyl dopa, rifampicin Autoimmune hemolytic anemia
5. Family history of anemia, jaundice, gallstones, Inherited hemolytic anemia
or splenectomy
6. Organomegaly (hepatomegaly, splenomegaly, Hematologic malignancy
lymphadenopathy)
7. Community:
• Sindhis, Lohanas Thalassemia
• Parsees, Vataliya Prajapatis Glucose 6 phosphate dehydrogenase deficiency
• Tribal groups Sickle cell disease
8. Deformities of skull and facial bones like Thalassemia
malar prominence, frontal bossing, depressed
bridge of nose
Laboratory Tests in Anemia 261

probable cause of anemia. Apart from morphological type Red cell indices: Among the red cell indices, mean cell
of anemia and abnormality of red cells, blood smear also volume is the most useful for classification of anemia
provides information regarding disorders of white cells into normocytic, microcytic, and macrocytic types
(e.g. leukemias), and platelets (thrombocytopenia). (See (Table 27.16). Red cell distribution width is used to
Chapter 22: Blood Smear). distinguish between iron deficiency anemia and
thalassemia (see Chapter 23: Red Cell Indices).
Reticulocyte count: It is a measure of the capacity of the
bone marrow to produce red cells. Reticulocyte count is
Microcytic Hypochromic Anemia
mainly useful for diagnosis of anemia due to decreased
red cell production or ineffective erythropoiesis (in which An approach for evaluation for microcytic hypochromic
reticulocyte count is low). Increased reticulocyte count anemia is shown in Figure 27.20. The distinction between
appropriate for degree of anemia indicates effective red different causes of microcytic anemia is shown in Table
cell production. Causes of low reticulocyte count are 27.3 earlier.
megaloblastic anemia, aplastic anemia, bone marrow
infiltration, and alcoholism. Causes of increased Macrocytic Anemias
reticulocyte count are response to hematinic therapy in Evaluation of macrocytic anemia is presented in Figure
nutritional anemia, hemolysis, and bleeding (see Chapter 27.21.
21: Reticulocyte Count). If reticulocyte count is increased,
then tests for hypoproliferative anemias are generally not Normocytic Normochromic Anemia
required (such as iron studies, vitamin B12 and folate Evaluation of normocytic anemia is shown in Figure
assays, bone marrow examination). 27.22.

Table 27.16: Morphological classification of anemia


Microcytic hypochromic anemia
(MCV < 80 fl; MCHC < 30 gm/dl)
• Iron deficiency anemia
• Thalassemia
• Anemia of chronic disease
• Sideroblastic anemia
Macrocytic anemia*
(MCV > 100 fl; MCHC 30-35 gm/dl)
• Megaloblastic • Non-megaloblastic
– Folate or vitamin B12 deficiency – Alcoholism
– Liver disease
– Hemolytic anemia
– Myelodysplastic syndrome
– Hypothyroidism

Normocytic and normochromic anemia


(MCV 80-100 fl; MCHC 30-35 gm/dl)
• Reticulocyte count increased • Reticulocyte count low
– Recent blood loss – Aplastic anemia
– Hemolysis – Chronic renal failure
– Anemia of chronic disease
– Anemia due to infiltration of marrow

*Another way of classification of macrocytic anemia: (1) Round macrocytosis: alcoholism, liver disease, hypothyroidism;
and (2) oval macrocytosis: folate or vitamin B12 deficiency, myelodysplastic syndrome. Abbreviations: MCV: mean cell
volume; MCHC: mean cell hemoglobin concentration
262 Essentials of Clinical Pathology

Fig. 27.20: Evaluation of microcytic hypochromic anemia

Fig. 27.21: Evaluation of macrocytic anemia


Laboratory Tests in Anemia 263

Fig. 27.22: Evaluation of normocytic normochromic anemia

Hemolytic Anemia Approach to the diagnosis of hemolytic anemia


Increased rate of red cell destruction which cannot be consists of determining (i) presence of hemolysis, (ii)
compensated by increased red cell production by the whether hemolysis is intravascular or extravascular, and
bone marrow leads to hemolytic anemia. Normally, red (iii) cause of hemolysis.
cells have a life span of about 120 days after which they 1. Establishing the presence of hemolysis: Laboratory
are destroyed. In hemolytic anemia, red cell life-span is features of excessive red cell destruction are:
shortened. Causes of hemolytic anemia are listed earlier • Increased indirect serum bilirubin
in Table 27.1. • Increased urobilinogen in urine
Apart from general features of anemia, hemolytic • Decreased or absent plasma haptoglobin (protein
anemia may manifest clinically with jaundice, and in that binds free hemoglobin in plasma)
chronic cases with gallstones and splenomegaly. Family • Appearance of free hemoglobin in plasma or urine
history may be positive in hereditary hemolytic anemias, • Increased serum lactate dehydrogenase
and occurrence in a particular ethnic group may be
suggestive of a particular disorder (e.g. thalassemia, Laboratory features of increased red cell production by
sickle cell disease, glucose-6-phosphate dehydrogenase the bone marrow (in response to hemolysis) are:
deficiency). In acquired hemolytic anemias, evidence of • Increased reticulocyte count
an underlying disorder may be present, e.g. malaria, • Polychromatic cells and nucleated red cells on blood
septicemia, drug ingestion, lymphoproliferative disorder, smear
connective tissue disease, disseminated cancer, etc. • Erythroid hyperplasia in bone marrow.
264 Essentials of Clinical Pathology

The three typical laboratory features indicative of In extravascular hemolysis, unconjugated bilirubin in
hemolytic anemia are low hemoglobin, increased serum and urobilinogen in urine are increased. However,
reticulocyte count, and raised indirect serum bilirubin. hemoglobinemia, hemoglobinuria, hemosiderinuria, and
2. Whether hemolysis is intravascular or extravascular: In methemalbuminemia are absent.
intravascular hemolysis, red cell destruction occurs 3. Defining cause of hemolysis: Usually, clinical features
within circulation (Fig. 27.23), while in extravascular and morphology of red cells on blood smear suggest
hemolysis, red cells are destroyed by macrophages the probable cause of hemolytic anemia (Fig. 27.25).
of the reticuloendothelial system in spleen and liver Specific laboratory tests are then carried out for
(Fig. 27.24). definitive diagnosis.
Main causes of intravascular hemolysis are hemolytic Specific laboratory studies to establish the cause of
transfusion reaction, glucose-6-phosphate dehydro- hemolytic anemia are:
genase deficiency, blackwater fever in falciparum
malaria, septicemia, autoimmune hemolytic anemia a. Tests in hereditary hemolytic anemias:
(some types), and paroxysmal nocturnal hemoglobinuria. • Hemoglobin disorders: sickling test, hemoglobin
Laboratory features of intravascular hemolysis are: electrophoresis, high performance liquid
• Hemoglobinemia chromatography, estimation of hemoglobin A2,
• Decreased or absent plasma haptoglobin estimation of hemoglobin F, red cell inclusions.
• Hemosiderinuria • Red cell enzyme defects: test for glucose-6-
• Methemalbuminemia: Methemalbumin has a phosphate dehydrogenase deficiency
characteristic absorption band at 558 nm (Schumm’s • Red cell membrane disorders: osmotic fragility
test). test, autohemolysis test

Fig. 27.23: Catabolism of hemoglobin following intravascular hemolysis.


Laboratory Tests in Anemia 265

b. Tests in acquired hemolytic anemias:


• Immune hemolytic anemias: antiglobulin
(Coombs’) test
• Paroxysmal nocturnal hemoglobinuria: sucrose
hemolysis test, Ham’s acidified serum lysis test,
flow cytometric analysis.

Specific Laboratory Studies in Anemia

Vitamin B12 and Folate


Vitamin B12 and folate are measured in patients with (i)
MCV>100 fl (macrocytosis), (ii) macrocytic anemia, or
(iii) neurological and psychiatric abnormalities. Some
investigators also recommend measurement of serum
methylmalonic acid and serum homocysteine due to
interpretive difficulties associated with vitamin assays.

Iron Studies

Iron studies for diagnosis of iron deficiency anemia


include serum iron, total iron binding capacity,
transferrin saturation, and serum ferritin. Before testing,
causes of iron deficiency and presence of any chronic
disease should be assessed. Serum ferritin is superior to
other tests for diagnosis of iron deficiency (< 15 μg/dl is
Fig. 27.24: Catabolism of hemoglobin following diagnostic of iron deficiency, while >100 μg/dl excludes
extravascular hemolysis iron deficiency).

Fig. 27.25: Evaluation of hemolytic anemia


266 Essentials of Clinical Pathology

Bone Marrow Examination A positive test indicates presence of Hb S. The


test cannot differentiate between sickle cell trait and
If facilities for iron studies and vitamin assays are not sickle cell disease. However, if the test is positive, then
available, examination of bone marrow is indicated to on the basis of blood smear examination, a probable
(i) distinguish between causes of microcytic hypochromic diagnosis of sickle cell trait (target cells) or sickle cell
anemia by assessing storage iron and ringed sideroblasts, disease (presence of sickle cells and target cells) may
and (ii) distinguish between megaloblastic and non- be suggested. It is necessary to perform hemoglobin
megaloblastic macrocytosis. Bone marrow biopsy is electrophoresis for confirmation of Hb S and to
essential for diagnosis of aplastic anemia. differentiate between various sickle cell disorders
(sickle cell trait, sickle cell anemia, sickle cell-β
thalassemia, sickle cell-DPunjab disease, etc.).
Tests for Hemoglobin S
False-negative test can occur if the reagent is outdated
Two tests are available for detection of Hb S: or not freshly prepared, concentration of Hb S is low
• Sickle cell slide test (e.g. in infants below 6 months, following recent blood
transfusion), or if there is severe anemia.
• Solubility test for hemoglobin S
False-positive test can occur if the concentration of
1. Sickle cell slide test: When red cells containing Hb S
sodium metabisulphite is excessive or if there is
are deprived of oxygen, they become sickle-shaped. drying of the wet preparation.
Reducing agent that is used to remove oxygen from 2. Solubility test for hemoglobin S: In this test,
red cells is 2% sodium metabisulphite. anticoagulated blood is added to the reagent solution
A drop of capillary or anticoagulated venous blood consisting of phosphate buffer, saponin, and sodium
is mixed on a glass slide with a drop of 2% sodium dithionite. Red cells are hemolyzed and Hb S, if
metabisulphite. A coverslip is placed over the mixture present, is reduced by dithionite. Hb S forms tactoids
and sealed with petroleum jelly-paraffin wax. The which refract light and the solution appears turbid
preparation is examined under the microscope after (Fig. 27.27). The solution remains clear with other
30 minutes. If sickle cells are not seen, examine the hemoglobins like Hb A, Hb F, Hb C, Hb D, Hb G,
slide again after 2 hours, and 24 hours. The test is and Hb O-Arab. The test cannot differentiate between
reported as negative if the red cells remain round, and various sickle cell disorders.
as positive if red cells become sickle shaped (crescent- Causes of false-positive test include polycythemia,
shaped with pointed ends) or holly-leaf shaped marked leukocytosis, paraproteinemias, and hyper-
(Fig. 27.26). lipidemia.

Fig. 27.27: Solubility test for sickle hemoglobin. In a negative


test, black lines kept behind the test tube are visible. In a positive
test, black lines are not visible because the solution becomes
Fig. 27.26: Sickle cell slide test turbid
Laboratory Tests in Anemia 267

Causes of false-negative test are old or outdated Cellulose acetate electrophoresis at alkaline pH is the
reagent, low concentration of Hb S (e.g. infants <6 first-line test in the investigation of hemoglobinopathies.
months, severe anemia), and recent blood transfusion. The procedure in short is as follows:
If the test is positive, hemoglobin electrophoresis 1. Hemolysates are prepared from EDTA antico-
should be performed for confirmation of Hb S, and to agulated blood (both control and patient’s samples)
distinguish between various sickle cell disorders. to obtain hemoglobin solutions. The control consists
of hemoglobins A, F, S, and C.
Hemoglobin Electrophoresis 2. Samples are applied near one end of the cellulose
There are different forms of normal (e.g. HbA, Hb F, Hb acetate strip (point of origin) in separate lanes.
A 2 ) and abnormal (e.g. Hb S, Hb C, Hb D, Hb G) 3. Cellulose acetate strips are placed in the electro-
hemoglobins. The proportions of different hemoglobins phoresis chamber containing the Tris-EDTA-borate
in common disorders of hemoglobin are shown in Table buffer (pH 8.5) with point of origin towards the
27.17. Various hemoglobins differ in their amino acid cathode.
composition, and hence in their charge when placed in 4. The chamber is connected to the power supply and
an electric field and migrate at different rates. Electro- electric current is applied till adequate separation of
phoresis can detect only those abnormalities of hemo- hemoglobins is obtained.
globin that alter the charge. These hemoglobins are 5. The strips are removed from the chamber and results
identified by positions they occupy when they migrate are read visually. If a permanent record is required,
from their point of origin. A control sample consisting strips are stained with a protein stain.
of known normal and abnormal hemoglobins is also run
Relative mobilities of some hemoglobins after
along with the test sample so as to compare and identify
electrophoresis at alkaline pH are shown in Figure 27.28.
locations of hemoglobins in the test sample.
Hemoglobin electrophoresis is used for: At alkaline pH, some hemoglobins have the same net
• Detection of abnormal hemoglobins like Hb S, Hb C, charge so that they migrate to the same position, e.g.
Hb D, Hb G, etc. and determining the phenotype by hemoglobins S, D, and G occupy the same position, and
assessing relative proportions of Hb A, Hb F, and the A2 , C, E, and O run together. To differentiate these
abnormal hemoglobin. hemoglobins from each other, citrate agar electrophoresis
• Detection of elevated HbF or Hb A2 in thalassemias. should next be carried out at acid pH (see Fig. 27.28).
• Quantitation of Hb A2 for diagnosis of β thalassemia Citrate agar electrophoresis at acid pH (6.0) provides
trait. separation of hemoglobins that migrate to the same
Two types of hemoglobin electrophoresis are commonly position on cellulose acetate at pH 8.5. In addition, it is
used: also well-suited for neonatal screening of sickle cell
• Cellulose acetate electrophoresis at alkaline pH anemia since clear separation between hemoglobins A,
• Citrate agar electrophoresis at acid pH. F, and S is obtained.

Table 27.17: Proportions of different hemoglobins in normal individuals and in hemoglobin disorders
Condition HbA HbF HbA2 HbS

1. Normal adults 97% <1% 1-3% 0


2. Normal newborn 25% 75% <1% 0
3. Sickle cell trait 56-60% 0 1-3% 40%
4. Sickle cell anemia 0 5-10% 1-3% 90-95%
5. β thalassemia minor 90-95% 0-5% 3.5-7% 0
6. β thalassemia major
0
0 95-98% 2-5% 0
268 Essentials of Clinical Pathology

Fig. 27.28: Diagrammatic representation of hemoglobin electrophoresis


(Control is a mixture of hemoglobins A, F, C, and S)

High Performance Liquid Chromatography Estimation of Hemoglobin A2 (Hb A2)


In this chromatographic technique, blood sample is Measurement of HbA2 is carried out for diagnosis of β
introduced in a matrix column. Mixture of hemoglobins thalassemia carriers; in these cases HbA2 is characteris-
gets adsorbed onto the matrix and each hemoglobin is tically raised to 3.5-7.0%.
eluted at a different time. Absorbance of eluted There are two methods for estimation of Hb A2:
hemoglobins is measured (Fig. 27.29). • Cellulose acetate electrophoresis at alkaline pH
followed by elution and comparing absorbance of Hb
A2 eluate against absorbance of eluates from other
hemoglobin bands.
• High performance liquid chromatography.

Estimation of Hemoglobin F (Hb F)


Fetal hemoglobin (Hb F) is the predominant form of
hemoglobin during fetal life, and at birth constitutes 75%
of all hemoglobins. After birth, Hb F gradually declines
and is about 5% at 6 months and less than 2% at 1 year.
In adults, Hb F is about 0.5%.
Markedly raised Hb F level occurs in:
• β thalassemia major
• Hereditary persistence of fetal hemoglobin (a form
of mild thalassemia)
• Sickle cell diseases.
Hb F is commonly measured by alkali denaturation
test.

Red Cell Inclusions


Fig. 27.29: Diagrammatic representation of chromatograms In thalassemias, red cell inclusions can be demonstrated
obtained with high performance liquid chromatography by staining with methyl violet. Hb H inclusions represent
Laboratory Tests in Anemia 269

Fig. 27.31: Osmotic fragility test

Fig. 27.30: Heinz bodies in red cells in glucose 6


phosphate dehydrogenase deficiency

precipitated tetramers of β globin chains in red cells in α


thalassemias. They appear as multiple, small, ragged
inclusions. α chain inclusions are seen in β thalassemia in
erythroblasts in bone marrow and appear as single
ragged structures closely attached to the nucleus.
Heinz bodies represent precipitated denatured
hemoglobin and are seen in unstable hemoglobin disease
and glucose-6-phosphate dehydrogenase deficiency.
They are deep purple and attached to the cell membrane
(Fig. 27.30).

Osmotic Fragility Test


Fig. 27.32: Osmotic fragility curve. Percent lysis is plotted on
In this test, red cells are suspended in decreasing
vertical axis and concentration of saline is plotted on horizontal
concentrations of hypotonic saline solutions to determine axis. In hereditary spherocytosis (red), curve is shifted to the
the ability of the red cells to withstand osmotic stress. In right of the normal, while in thalassemia (blue), curve is shifted
hypotonic solutions, water enters red cells causing to the left of the normal
cellular swelling, and at one point, cell lysis occurs.
Normal red cells are biconcave and disc-shaped, have In the presence of spherocytosis, red cells show
high surface area to volume ratio, and therefore can beginning of hemolysis at 0.6 to 0.8 gm/dl (i.e. increased
increase their volume upto 70% before they are lysed. osmotic fragility). Increased number of spherocytes (and
Spherocytes have decreased surface area to volume ratio consequently increased osmotic fragility) is seen in
and therefore they can take up less water than normal hereditary spherocytosis, warm antibody type auto-
red cells and lyse earlier (i.e. at relatively higher saline immune hemolytic anemia, ABO hemolytic disease of
concentration than normal red cells). newborn, and burns.
With normal red cells, hemolysis usually starts at Osmotic fragility is decreased in the presence of target
saline concentration of 0.5 gm/dl and is complete at 0.30 cells (e.g. in thalassemia) since target cells have increased
gm/dl (Figs 27.31 and 27.32). surface volume.
270 Essentials of Clinical Pathology

This test may be negative if only a few spherocytes are Antiglobulin (Coombs’) Test
present in peripheral blood. Sensitivity of the test can be
Antiglobulin or Coomb’s test is of two types: direct
increased by incubating the red cells at 37°C for 24 hours
(Fig. 27.34) and indirect (Fig. 27.35).
before performing the test (osmotic fragility test after
incubation). Direct antiglobulin test (DAT): This test detects antibodies
or complement or both attached to red cells. DAT is used
Autohemolysis Test
in the investigation of acquired immune hemolytic
In autohemolysis test, blood is incubated at 370 C for 48
hours and the amount of spontaneous hemolysis is noted.
The test is carried out with and without addition of
glucose. In hereditary spherocytosis, autohemolysis is
markedly increased, which is corrected partially
following addition of glucose (Fig. 27.33).

Tests for Glucose-6-Phosphate


Dehydrogenase Deficiency
Definitive diagnosis depends on demonstration of G6PD
deficiency by fluorescent spot test, methemoglobin
reduction test, or dye decolorization test.
Recommended test is fluorescent spot test. G6PD is
required for conversion of NADP to NADPH. Under
ultraviolet light, NADPH fluoresces while NADP does
not. In the absence of G6PD, NADPH will not form and
there will be no fluorescence. Presence of fluorescence
indicates normal G6PD activity.

Fig. 27.34: Principle of direct antiglobulin (Coombs’) test

Fig. 27.33: Autohemolysis test. Left two tubes are control


samples. Glucose has been added to the second normal tube
leading to partial correction of hemolysis. Two tubes on the
right are test samples from a patient with hereditary sphero-
cytosis. The first tube from the patient is showing marked
hemolysis. After addition of glucose, hemolysis is partially
corrected Fig. 27.35: Principle of indirect antiglobulin test
Laboratory Tests in Anemia 271

anemia and determines whether hemolysis is of immune Applications of IAT


type. In principle, red cells of the patient are washed with 1. Cross matching (compatibility testing) before blood
normal saline (to remove unbound antibodies) and transfusion.
antihuman globulin (AHG) reagent is added. 2. Antibody screening and identification.
Agglutination of red cells indicates positive test. (AHG 3. Phenotyping of red cell antigens using known
sera are either polyspecific, i.e. they contain antibodies to antisera.
both human IgG and C3d, or monospecific, i.e. they contain 4. Titration of anti-Rh antibodies.
antibodies to either IgG or C3b-C3d).
Tests for Paroxysmal Nocturnal Hemoglobinuria
Causes of positive test:
1. Sucrose hemolysis test: This is the screening test. It is
1. Autoimmune hemolytic anemia (coating of red cells
highly sensitive but less specific than Ham’s test. Red
with autoantibodies). cells from the patient are mixed with fresh ABO-
2. Hemolytic disease of newborn (coating of red cells of compatible normal serum and isotonic sucrose
fetus with maternal antibodies). solution (a low ionic strength medium which activates
3. Hemolytic transfusion reaction (coating of red cells complement). Hemolysis >10% indicates PNH.
of donor with recipient antibodies). 2. Ham’s acidified serum lysis test: Acidified serum lysis
4. Drug-induced hemolysis, e.g. methyldopa, L-dopa, test or Ham’s test is the definitive test for the diagnosis
cephalothin, penicillin, etc. of PNH. Patient’s red cells are mixed with acidified
normal serum (acidification activates complement).
Indirect antiglobulin test (IAT): This test detects presence Hemolysis occurs if red cells are abnormally sensitive
of antibodies in serum which are directed against red to complement action (Fig. 27.36). This test may also
cell antigens. In this test, patient’s serum is incubated be positive in a rare form of congenital
with red cells (donor or screening) to allow binding of dyserythropoietic anemia called as hereditary
antibodies in serum to red cell antigens. After washing erythroblast multinuclearity with positive acidified
of red cells in saline (to remove unbound antibodies), serum. Examination of bone marrow for abnormal
antihuman globulin reagent is added. Agglutination of erythroblasts can differentiate between these two
red cells denotes positive test. conditions.

Fig. 27.36: Ham’s acidified serum lysis test for paroxysmal nocturnal hemoglobinuria. Tube 1: Fresh normal serum + Patient’s
red cells; Tube 2: Fresh normal serum + Hydrochloric acid + Patient’s red cells; Tube 3: Patient’s serum + Hydrochloric acid
+ Patient’s red cells; Tube 4: Heat-inactivated normal serum + Hydrochloric acid + Patient’s red cells; Tube 5: Fresh normal
serum + Normal red cells; Tube 6: Fresh normal serum + Hydrochloric acid + Normal red cells; Tube 7: Heat-inactivated
normal serum + Hydrochloric acid + Normal red cells. Tube 1 is showing trace hemolysis, tube 2: marked hemolysis;
tube 3: slight hemolysis; tubes 4 to 7: No hemolysis
272 Essentials of Clinical Pathology

3. Flow cytometric analysis: This can detect deficiency of Vitamin B12 and Folate Studies
CD 55 or CD 59 proteins by using monoclonal • Serum vitamin B12: 150-700 ng/L
antibodies. • Serum folate: 3-20 µg/L
• Red cell folate: 150-700 µg/L
REFERENCE RANGES
Hemoglobin Serum Haptoglobin

• Adult males: 13.0-17.0 gm/dl. 0.8-2.7 g/L


• Adult females (non-pregnant): 12.0-15.0 gm/dl.
• Adult females (pregnant): 11.0-14.0 gm/dl. CRITICAL VALUES
• Children, 6-12 years: 11.5-15.5 gm/dl. • Hemoglobin <7 g/dl or >20 g/dl
• Children, 6 months to 6 years: 11.0-14.0 gm/dl. • Packed cell volume <20% or >60%
• Infants, 2-6 months: 9.5-14.0 gm/dl. • Presence of sickle cells on blood smear
• At birth (full term): 13.6-19.6 gm/dl. • New diagnosis of aplastic anemia

Packed Cell Volume BIBLIOGRAPHY


1. Bacigalupo A, Hows JM, Gluckman E, Nissen C, Marsh
• Adult males: 40-50% J, Van Lint MT, Congiu M, De Planque MM, Ernst P,
• Adult females (non-pregnant): 38-45%. McCann S. Bone marrow transplantation (BMT) versus
• Adult females (pregnant): 36-42% immunosuppression for the treatment of severe aplastic
• Children, 6-12 years: 37-46% anemia (SAA): a report of the EBMT SAA Working
• Children, 6 months to 6 years: 36-42% Party. Br J Haemat 1988;70:177-82.
2. Camitta BM, Thomas ED, Nathan DG, Santos G,
• Infants, 2 – 6 months: 32-42% Gordon-Smith EC, Gale RP, Rappeport JM, Storb R.
• At birth (full term): 44-60% Severe aplastic anemia: a prospective study of the effect
of early marrow transplantation on acute mortality.
Red Cell Count Blood 1976;48:63-9.
3. Clarke GM, Higgins TN. Laboratory investigation of
• Adult males: 4.5-5.5 million/cmm hemoglobinopathies and thalassemias: Review and
• Adult females: 3.8-4.8 million/cmm update. Clin Chem 2000;46:1284-90
4. Evatt BL, Gibbs WN, Lewis SM, McArthur JR. Funda-
Red Cell Indices mental diagnostic hematology. Anemia (2nd ed). US
Department of Health and Human Services, Georgia
• Mean cell volume (MCV): 80-100 fl and World Health Organization, Switzerland, 1992.
• Mean corpuscular hemoglobin (MCH): 27-32 pg 5. Kawthalkar SM. Essentials of Hematology. New Delhi:
• Mean corpuscular hemoglobin concentration Jaypee Brothers Medical Publishers (P) Ltd, 2006.
(MCHC): 32-36 g/dl 6. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical
Hematology. (9th Ed). London: Churchill Livingstone, 2001.
• Red cell distribution width: 9-14.5 7. Smellie WSA, Wilson D, McNulty CAM, et al. Best
practice in primary care pathology: review 1. J Clin
Osmotic Fragility of Red Cells Pathol 2005;58:1016-24.
• Starts at 0.5% of sodium chloride or lower 8. Tripathy V, Reddy BM. Present status of understanding
on the G6PD deficiency and natural selection. J Postgrad
• Complete at 0.3% of sodium chloride
Med 2007;53:193-202.
9. Ward PCJ. Modern approaches to the investigation of
Iron Studies vitamin B12 deficiency. Clin Lab Med 2002;22:435-45.
• Serum iron: 50-150 μg/dl 10. Working party of the BCSH Blood Transfusion and
General Hematology Task Forces: The estimation of
• Total iron binding capacity (TIBC): 300-400 μg/dl
fetomaternal hemorrhage. Transf Med 1999;9:87-92.
• Percent transferrin saturation: 20-55%
11. Working party of the General Hematology Task Force
• Free erythrocyte protoporphyrin (FEP): <80 μg/dl of the British Committee for Standards in Hematology:
• Serum transferrin receptor: 2.8-8.5 μg/L The laboratory diagnosis of hemoglobinopathies. Br J
• Serum ferritin: 15-300 μg/L Haemat 1998;101:783-92.
28
Laboratory Tests in
Hematological Malignancies

ACUTE LEUKEMIAS Clinically, patients with acute leukemia present with


(i) features due to suppression of normal hematopoiesis
Acute leukemias are malignant clonal hematopoietic like anemia (pallor, weakness, fatigue), neutropenia
stem cell disorders characterized by proliferation of blast (infections), and thrombocytopenia (purpura and
cells in the bone marrow and rapidly progressive fatal mucosal bleeding), (ii) features due to accumulation of
course if untreated. Malignant cells in acute leukemias blast cells in marrow (bony pain and tenderness), and
are primitive cells with very little differentiation into (iii) features due to infiltration of various organs by blast
functioning mature cells and are called as blast cells. cells (lymphadenopathy, splenomegaly, hepatomegaly,
Acute leukemias are primary disorders of the bone swelling of gums, central nervous system manifestations,
marrow and arise from malignant transformation of a and skin infiltrations).
single hematopoietic progenitor cell followed by Classification of Acute Leukemias
proliferation and accumulation of the abnormal clone.
The most widely used classification of acute leukemias
Proliferating leukemic blast cells replace normal bone is French-American-British (FAB) Co-operative Group
marrow and subsequently enter into the peripheral classification, which was originally proposed in 1976.
blood. In the bone marrow, blast cells inhibit the growth This classification is based on morphology and cyto-
and proliferation of normal hematopoietic cells. After chemistry. Acute leukemias are divided into two major
entering the peripheral blood, leukemic cells can involve types: acute myeloid leukemia (AML) and acute
any organ system but lymph nodes, liver, spleen, central lymphoblastic leukemia (ALL). Each type is further
nervous system, and skin are commonly affected. subclassified (Table 28.1).

Table 28.1: French-American-British (FAB) classification of acute leukemias

Acute myeloid leukemia (AML)


M0: Acute myeloid leukemia, minimally differentiated
M1: Acute myeloid leukemia without maturation
M2: Acute myeloid leukemia, with maturation
M3: Acute promyelocytic leukemia M3v: Hypo- or microgranular promyelocytic leukemia
M4: Acute myelomonocytic leukemia M4Eo: Acute myelomonocytic leukemia with bone marrow eosinophilia
M5: Acute monocytic leukemia M5a: Undifferentiated (monoblastic) M5b: Well-differentiated (promonocytic-monocytic)
M6: Acute erythroleukemia
M7: Acute megakaryocytic leukemia

Acute lymphoblastic leukemia (ALL)


L1
L2
L3
274 Essentials of Clinical Pathology

Recently, World Health Organization (WHO) classifi- for diagnosis of AML (Earlier FAB recommendation was
cation of hematopoietic and lymphoid neoplasms has 30%). The term ‘blast’ includes myeloblasts, promyelo-
been proposed (2001). WHO classification (2001) of acute cytes (for acute promyelocytic leukemia), monoblasts and
myeloid leukemias is shown in Table 28.2. This promonocytes (for acute myelomonocytic and acute
classification integrates clinical, morphologic, genetic, and monocytic leukemia), and megakaryoblasts (for acute
immunophenotypic features to define specific disease megakaryocytic leukemia).
categories. Myeloid cell lines are erythroid, granulocytic,
In WHO classification, acute lymphoblastic leukemias monocytic, and megakaryocytic. Myeloid nature of blasts
are placed under lymphoid neoplasms. ALL and is established by morphology (Auer rods), cytochemistry
lymphoblastic lymphoma or Burkitt’s lymphoma are
(myeloperoxidase, nonspecific esterase), and immuno-
considered as biologically the same disease with different
phenotyping (myeloid related antigens like CD 117, CD
clinical presentations. The term ALL is used for leukemic
13, CD 33).
phase of precursor neoplasms of B or T cell type. Burkitt
In contrast to FAB classification, WHO classification
cell leukemia is the counterpart of Burkitt lymphoma and
recognizes AML with recurrent cytogenetic abnormalities,
corresponds with FAB L3 subtype of ALL. ALL includes
following categories in WHO classification: AML with multilineage dysplasia, and therapy-related
• Precursor B cell ALL AML as distinct entities.
• Precursor T cell ALL Patients with clonal, recurrent cytogenetic abnor-
• Burkitt cell leukemia malities listed in Table 28.2 are considered to have AML
Salient features of WHO classification are outlined in irrespective of the percentage of blasts in blood or bone
Table 28.2. marrow. These patients have characteristic clinical and
According to WHO criteria, blast count should be 20% morphological features and a favorable response to
or more in either peripheral blood smear or bone marrow therapy.

Table 28.2: WHO classification of acute myeloid leukemia (AML), 2001

1. Acute myeloid leukemia with recurrent genetic abnormalities


• AML with t (8; 21) (q22; q22), (AML1/ETO)
• AML with abnormal bone marrow eosinophils and inv (16) (p13q22) or t (16; 16) (p13; q22), (CBFβ /MYH11)
• Acute promyelocytic leukemia with t(15;17) (q22; q12), (PML/RARα ) and variants
• AML with 11q23 (MLL) abnormalities
2. Acute myeloid leukemia with multilineage dysplasia
• Following myelodysplastic syndrome (MDS)
• Without prior myelodysplastic syndrome
3. AML and MDS, therapy related
• Alkylating agent related
• Topoisomerase II inhibitor related
• Others
4. AML, not otherwise categorized
• AML, minimally differentiated
• AML without maturation
• AML with maturation
• Acute myelomonocytic leukemia
• Acute monoblastic and monocytic leukemia
• Acute erythroid leukemia
• Acute megakaryoblastic leukemia
• Acute basophilic leukemia
• Acute panmyelosis with myelofibrosis
• Myeloid sarcoma
Laboratory Tests in Hematological Malignancies 275

Diagnosis of AML with multilineage dysplasia 1. Morphology: Initial step in the diagnosis of acute
requires either (i) prior history of myelodysplastic leukemias is examination of Romanowsky-stained
syndrome for atleast 6 months before the onset of overt smears of peripheral blood and marrow aspirate.
AML, or (ii) ≥ 50% of dysplastic cells in two or more In a typical case, bone marrow suppression leads
myeloid lineages. This category of AML is associated to normocytic normochromic anemia, thrombo-
with poor outcome. cytopenia, and neutropenia. Total leukocyte count is
Features of alkylating therapy related AML are (i) usually elevated; however, it may be normal or low.
development 4-7 years following therapy, (ii) charac- In subleukemic leukemia, leukocyte count is normal or
teristic cytogenetic abnormalities involving chromo- decreased, but blasts are demonstrable in peripheral
somes 5 or 7, (iii) associated and preceding multilineage blood. In aleukemic leukemia, blasts are not demonst-
dysplasia, and (iv) adverse prognosis. rable in peripheral blood, but present in bone marrow.
Topoisomerase II inhibitor therapy-related AML In AML (M0, M1), leukemic hiatus occurs, i.e.
develops 6 months to 5 years after starting therapy, and peripheral blood shows blast cells and mature cells,
is also associated with characteristic cytogenetic while intermediate cells (like promyelocytes,
alterations affecting 11q23. There is no preceding myelocytes, and metamyelocytes) are not seen.
myelodysplasia. This form of therapy can also cause acute Bone marrow is usually hypercellular due to
lymphoblastic leukemia. infiltration by monomorphic population of leukemic
Cases of AML which do not fit into any of the above cells; normal hematopoietic cells are reduced.
three major categories are placed under the category of Blast percentage should be derived from 200-cell
AML, not otherwise categorized. differential leukocyte count on a blood smear or 500-
According to WHO classification, FAB terms L1, L2, cell differential count on a marrow aspirate smear.
and L3 are not distinct disease categories since they do Presence of Auer rods (needle- or rod-shaped
not have clinical, immunophenotypic, or genetic inclusion in cytoplasm composed of azurophil
correlates, and therefore these terms should no longer granules) in blast cells is diagnostic of AML.
be used. L3 subtype of ALL, however, correlates with However, they are seen in only 10-20% cases of AML.
Burkitt’s lymphoma. Morphological features of ALL and AML are shown
In WHO classification, the term ALL corresponds in Tables 28.3 and 28.4 and Figures 28.1 and 28.2.
with leukemic phase of precursor neoplasms of B or T Features of AML and ALL are compared in Table 28.5.
lymphocytic lineage. For diagnosis of ALL, blast count 2. Cytochemistry: Cytochemistry comprises of techni-
in marrow should be greater than 25%. ques for identification of enzymes, fats, or certain
other substances in the cytoplasm of blood cells. In
Laboratory Diagnosis of Acute Leukemias acute leukemia, cytochemistry is mainly useful for
Currently, diagnosis of leukemias is based on combi- identifying various subtypes of AML. In lymphoid
nation of clinical features, microscopic examination of leukemias, cytochemistry has been replaced by
peripheral blood and bone marrow, cytochemistry, immunophenotyping for characterization of different
immunophenotyping by flow cytometry, cytogenetics, subtypes. The results of cytochemistry should always
and molecular analysis. be interpreted along with conventional morphology

Table 28.3: Morphological features of ALL (FAB classification)


FAB classification Morphological features
1. ALL L1 Predominantly small cells with scanty cytoplasm; nuclear membrane
regular; nucleoli indistinct
2. ALL L2 Large cells, heterogeneous in size with moderate amount of cytoplasm;
nuclear membrane is irregular with clefting; nucleoli 1-2 and prominent
3. ALL L3 Large cells with moderate amount of deeply basophilic cytoplasm;
prominent cytoplasmic vacuoles; regular nuclear membrane; 1-2
prominent nucleoli
276 Essentials of Clinical Pathology

Table 28.4: Laboratory features of acute myeloid leukemias

Type Morphology Cytochemistry Immunophenotyping Other features

1. AML M0 Large, agranular blasts MPO – (+ on EM) CD13+, CD 33+, –


CD34+, HLADR+
2. AML M1 Minimal maturation (≥ 90% MPO + (>3% CD13+, CD33+, CD34+, t (9;22)
type I and II blasts in marrow); blasts) HLADR+
remaining cells maturing
granulocytes or monocytes
3. AML M2 20-89% non-erythroid cells MPO+ CD13+, CD33+, t(8;21)
are myeloblasts; type II blasts HLADR+
common; Auer rods frequent;
full range of granulocyte
maturation
4. AML M3 Abnormal promyelocytes MPO+ CD13+, CD33+ t(15;17),
predominant (>50%); in (strong) disseminated
hypergranular type, nucleus is intravascular
reniform and there are abundant coagulation
cytoplasmic granules and common
numerous Auer rods; in
microgranular variant, granules
are small and few, nucleus is
convoluted, and TLC is very high
5. AML M4 Both myeloblasts and monoblasts MPO+ ≥20% CD13+, CD33+, Inv(16) in M4
are ≥20% of nonerythroid cells blasts; CD14+, CD64+, with eosinophilia;
NSE+ ≥20% cells HLADR+ increased lysozyme
in blood and urine
6. AML M5 >80% cells monocytic NSE+ CD14+, CD64+, t(9;11)
HLADR+
7. AML M6 >20% of nonerythroid cells are PAS+ Glycophorin A+, CD71+
myeloblasts; >50 of all nucleated
cells are erythroblasts; marked
dyserythropoiesis
8. AML M7 >20% of blasts that may be Platelet CD41+, CD61+,
small, lymphoblast-like or large peroxidase+ HLADR+
with fine chromatin and prominent (EM)
nucleoli and cytoplasmic blebs

MPO: Myeloperoxidase; EM: Electron microscopy; Type I blasts: No azurophil granules in cytoplasm; Type II blasts: Few
azurophil granules; NSE: Non-specific esterase; PAS: Periodic Acid Schiff; TLC: Total leucocyte count

and immunophenotyping. The important cyto- • MPO stain is positive in AML M1, AML M2, AML M3,
chemical studies in acute leukemias are (Fig. 28.3): and AML M4 (only in myeloblasts). In AML M0, MPO
• Myeloperoxidase (identifies an enzyme present activity is visible only by electron microscopy.
in primary granules of granulocytic cells) • Lymphoblasts are negative for MPO.
• Non-specific esterase (identifies an enzyme
present in large amounts in monocytic cells)
• Periodic acid Schiff (identifies intracellular Nonspecific Esterase (NSE)
glycogen).
NSE activity is strongly positive in monocytic series
Myeloperoxidase (MPO) (monoblasts, promonocytes, and monocytes). It allows
• MPO enzyme is located in primary (azurophil) and diagnosis of AML M4 and AML M5 (Fig. 28.5).
secondary granules in all stages of neutrophil series Sodium fluoride may be added that renders monocytic
(Fig. 28.4). cells negative.
Laboratory Tests in Hematological Malignancies 277

Fig. 28.1: (A) AML M0; (B) AML M1; (C) AML M2; (D) AML M3; (E) AML M4; (F) AML M5; (G) AML M6; (H) AML M7

Fig. 28.2: (A) ALL L1; (B) ALL L2; (C) ALL L3
278 Essentials of Clinical Pathology

Table 28.5: Differences between acute lymphoblastic leukemia and acute myeloid leukemia
Parameter Acute lymphoblastic leukemia Acute myeloid leukemia
1. Predominant age Children (peak 3-4 years) Adults
2. Morphology of blasts
• Size of blasts Small Large
• Cytoplasm Scanty Moderate
• Granules in cytoplasm Absent May be present
• Nuclear chromatin Coarse Fine
• Nucleoli 0-2 >3
• Auer rods Absent Pathognomonic, if present
3. Cytochemistry
• Myeloperoxidase Negative Positive
• Periodic acid Schiff Block-like Diffuse
4. Immunophenotyping B or T lymphoid markers Myeloid markers
5. Prognosis Curable in majority of children Curable in minority of adults

Fig. 28.3: Principle cytochemical reactions in acute leukemias

Periodic Acid Schiff (PAS) 3. Immunophenotyping: This technique consists of


• PAS reagent stains glycogen in the cytoplasm. identification of antigens present on leukemic cells
• Lymphoblasts show block-like positivity (Fig. 28.6). in blood or bone marrow with fluorescently-labeled
• PAS is positive in 70% of ALL L1 and ALL L2 cases. monoclonal antibodies. As blood and bone marrow
Lymphoblasts in ALL L3 are negative. cells are in fluid suspension, flow cytometery is the
method of choice. Cell surface antigens are named
Another stain that may be used is Sudan black B that dentifies according to the cluster of differentiation (CD) system.
phospholipids and other neutral fats in membranes of both Specific antigens are expressed on cells of different
primary and specific granules in granulocytes. lineages at different stages of development. A panel
Laboratory Tests in Hematological Malignancies 279

Fig. 28.4: Myeloperoxidase (MPO) stain. MPO is a marker


for primary azurophil granules and is the routine initial cyto- Fig. 28.6: Periodic acid Schiff reaction. Coarsely granular
chemical stain in all acute leukemias. Dark brown granules (‘block-like’) perinuclear staining pattern is observed in some
in the cytoplasm of blasts differentiate AML from ALL. However, cases of acute lymphoblastic leukemia. Negative PAS staining
negative MPO reaction does not exclude AML is not helpful for differentiating AML from ALL

helpful if morphology and cytochemistry are ambi-


guous. A large number of antigens can be assessed
simultaneously on a single specimen. The immuno-
logic profiles of cell lineages are follows:
• Primitive stem cell markers: CD34, CD117, TdT
• Myeloid lineage: CD13, CD33, cytoplasmic myelo-
peroxidase
• Monocytic lineage: CD14, CD64
• Erythroid lineage: Glycophorin A
• Megakaryocytic lineage: CD41, CD42, CD61
• B-ALL: CD19, CD20, CD21, CD22, CD79a, CD10
(common ALL antigen or CALLA), cytoplasmic
μ chain, surface immunoglobulin
• T-ALL: CD2, CD3, CD5, CD7, cytoplasmic CD3.
For further subclassification of B-ALL (into pro-B
ALL, common ALL, Pre-B ALL, mature B ALL) or T-
ALL (into pro-T ALL, pre-T ALL, cortical T ALL, mature
T ALL), another set of antibodies is needed.
Terminal deoxynucleotidyl transferase (TdT) is
Fig. 28.5: Nonspecific esterase reaction. This stain is used positive in all forms of ALL except L3 type.
for demonstration of monocytic differentiation in acute myeloid
Immunophenotyping is essential for diagnosis of
leukemia
AML M0 in which myeloperoxidase is negative on light
microscopy, myeloid-specific antigens (CD13, CD33,
consisting of a combination of specific antibodies is cytoplasmic myeloperoxidase, CD117) are positive, and
employed to determine the immunophenotype and T or B lymphoid markers are absent. CD14 and anti-
diagnostic classification of leukemias. It is especially lysozyme are strongly expressed in AML with significant
280 Essentials of Clinical Pathology

monocytic component (AML M4 and M5). In acute • Detection of minimal residual disease: Relapse
erythroleukemia (AML M6), glycophorin A and in acute following therapy is due to the persistence of viable
megakaryocytic leukemia (AML M7), platelet glyco- leukemic cells following cytotoxic chemotherapy.
protein antigens (CD41, CD42, and CD61) are demons- Leukemic cells on the background of normal cells can
trated. be detected by morphology, cytogenetics, immuno-
Immunophenotyping is also used for detection of phenotyping, and molecular methods.
minimal residual disease. Detection of minimal residual • To establish clonality i.e. determining whether a cell
disease (MRD) consists of identification of a small population is derived from a single cell (monoclonal)
population of leukemic cells among a large population or from multiple cells (polyclonal); it is helpful mainly
of normal cells after therapy to determine whether in B or T cell lymphomas.
residual disease is present that may later cause a relapse. • To detect chromosomal disorders which predispose
MRD can also be detected by morphology, cytogenetic to acute leukemia, e.g. trisomy 21.
analysis, and molecular analysis. 5. Molecular genetic analysis: This is used for:
4. Cytogenetic analysis: Structural or numerical • Detection of clonality by gene rearrangement
abnormalities of chromosomes are detected by studies (by polymerase chain reaction or Southern
cytogenetic analysis or karyotyping (Table 28.6). With blot analysis).
cytogenetic analysis, a variety of gross alterations can • Detection of minimal residual disease: Molecular
be detected such as translocations, deletions, and methods like polymerase chain reaction can be
duplications. In contrast to molecular methods, helpful in detecting a small submicroscopic
cytogenetic analysis is more widely available. In acute population of residual leukemic cells when
leukemias, cytogenetic abnormalities are linked to the peripheral blood and bone marrow examinations
pathogenesis of the disease. appear normal.
Applications of cytogenetic analysis in acute leukemia are • Diagnosis of specific types of acute leukemias by
• Diagnosis of specific types of AML: WHO classi- specific molecular probes: Molecular methods are
fication of AML recognizes distinct entities, which used for detection of chromosomal translocations
have clonal, recurrent cytogenetic abnormalities. that generate fusion transcripts and chimeric
Their identification is necessary as they have a proteins. The commonly used methods are reverse
correlation with response to therapy. transcription-polymerase chain reaction (RT-PCR)
• Prediction of prognosis: Certain cytogenetic abnor- and fluorescent in situ hybridization (FISH). The
malities are associated with poor prognosis and their technique detects t(15;17) in acute promyelocytic
detection may affect treatment strategies. leukemia that generates PML/RARα fusion gene,
and t(9;22) in B-ALL that generates bcr/abl fusion
Table 28.6: Chromosomal abnormalities in gene. Detection of these translocations is also
acute leukemias helpful for determining prognosis and response
Chromosomal abnormality Prognosis to treatment.
• Detection of opportunistic pathogens in immuno-
Acute lymphoblastic leukemia compromised patients.
1. t(9;22)(q34;q11.2) Poor
2. t(4;11)(q21;q23) Poor
CHRONIC LEUKEMIAS
3. t(1;19)(q23;p13.3) Average
4. t(12;21)(p13;q22) Good
Chronic leukemias are heterogeneous disorders
5. Hyperdiploidy>50 Good
6. Hypodiploidy Poor characterized by neoplastic proliferation of mature-
looking cells of myeloid or lymphoid lineage. General
Acute myeloid leukemia differences between acute and chronic leukemias are
1. t(8;21)(q22;q22) Good outlined in Table 28.7.
2. t(15;17)(q22;q12) Good Chronic leukemias are of two main types:
3. inv(16) Good
• Chronic myeloid leukemia
4. Monosomy 7 Poor
• Chronic lymphoid leukemias (Table 28.8).
Laboratory Tests in Hematological Malignancies 281

Table 28.7: General differences between acute and chronic leukemias


Parameter Acute leukemias Chronic leukemias
Clinical presentation Usually sudden and fulminant Usually incidental or insidious onset
Hematological features in Immature blast cells Mature and differentiated cells
marrow and blood
Course Aggressive Indolent

Table 28.8: Chronic lymphoid leukemias


B cell type T cell type
1. Chronic lymphocytic leukemia 1. Prolymphocytic leukemia
2. Prolymphocytic leukemia 2. Large granular lymphocytic leukemia
3. Waldenström macroglobulinemia 3. Adult T cell leukemia/lymphoma
4. Hairy cell leukemia 4. Sezary syndrome
5. Plasma cell leukemia
6. Leukemic phase of non-Hodgkin’s lymphoma

Chronic Myeloid Leukemia 22. The chimeric gene bcr-abl codes a tyrosine kinase that
affects cell proliferation.
Chronic myeloid leukemia (CML) is a chronic myelo-
proliferative neoplasm originating from a pleuripotent Chronic phase: The average age at diagnosis is 45 years. In
hematopoietic stem cell and characterized by predomi- chronic phase, the usual presenting features include
nant proliferation of granulocytic cells. weakness, weight loss, abdominal fullness, easy
There are three phases of CML: chronic phase (3-5 bruisability, and splenomegaly.
years), accelerated phase (6-12 months), and blast crisis Blood smear in chronic phase of CML shows marked
(2-4 months). leukocytosis, immature white blood cells, basophilia,
CML is associated with a characteristic cytogenetic eosinophilia, anemia, and thrombocytosis (Fig. 28.8).
abnormality called Philadelphia chromosome that results
from t(9;22)(q34;q11) (Fig. 28.7). This leads to juxtaposition
of c-abl gene from chromosome 9 to bcr gene on chromosome

Fig. 28.8: Blood smear in chronic phase of chronic myeloid


Fig. 28.7: Translocation between chromosomes 9 and 22 leukemia showing all forms of immature white cells and a
causing formation of Philadelphia chromosome basophil
282 Essentials of Clinical Pathology

Box 28.1: Fluorescence in situ hybridization (FISH)


This is a combination of cytogenetic and molecular
techniques used to identify and localize the presence or
absence of specific chromosomes or chromosomal regions
through hybridization of fluorescent probes that bind
specifically to its complementary target sequence. Fluores-
cent microscope is used to detect the presence or absence
of fluorescent signals. FISH can be used in metaphase
chromosomes (FISH-metaphase) or in interphase cells
(FISH-interphase), and is a very versatile procedure.
The technique consists of (1) Preparation of fluorescent
probes: Probes are short sequences of single-stranded DNA
that are complementary to the portion of gene of interest;
Fig. 28.9: Diagrammatic representation of fluorescence in situ probes are labeled with a fluorescent dye, (2) Preparation
hybridization (FISH) in interphase nuclei. (A) Normal interphase of metaphase chromosomes or interphase cells that are fixed
nucleus showing two red dots (located on abl genes on two on a microscope slide, (3) Hybridization: Fluorescently-
chromosomes number 9) and two green dots (located on bcr labeled probe is applied to the denatured chromosomal DNA
genes on two chromosomes number 22). (B) Interphase and incubated. Attachment or hybridization occurs between
nucleus of a leukemic cell in chronic myeloid leukemia showing the probe and the complementary DNA sequence. Non-
one red dot (abl on normal chromosome 9), one green dot hybridized probes are removed by washing, (4) Detection:
(bcr on normal chromosome 22), and a combined dot on fused
This consists of observation of hybridization under fluorescent
bcr-abl gene on Philadelphia chromosome. This fusion causes
lighting. Fluorescent signals indicate hybridization or
a fluorescent color change (red+green = yellow) which is not
presence of complementary sequence of DNA, while absence
shown in the figure
of fluorescent signals indicates absence of complementary
sequence of DNA. Each probe can be labeled with a different
coloured fluorescent dye, so that a number of different probes
Bone marrow shows markedly increased myeloid to can be simultaneously detected in the same preparation.
erythroid ratio, < 10% blasts, and increased reticulin or In interphase FISH, probes are directly introduced into
fibrosis. Diagnostic feature is Philadelphia chromosome the cell. In contrast to conventional cytogenetic anlysis that
requires 7-10 days for detection of chromosomal abnor-
on cytogenetic analysis, and presence of a bcr-abl fusion malities, interphase FISH provides rapid diagnosis within
gene on fluorescence in situ hybridization (FISH) 1-2 days. Unlike, metaphase FISH, actual chromosomes
(Fig. 28.9). Principle of FISH is shown in Box 28.1. cannot be visualized with interphase FISH and the location
of abnormality on the chromosome cannot be seen.
Leukocyte alkaline phosphatase (LAP) score: This cytochemical
stain is used to demonstrate presence and amount of the
enzyme alkaline phosphatase within neutrophils. Blood
smear is prepared from finger stick, air-dried, fixed, and
stained. Enzyme activity is indicated by the presence of
bright blue granules in neutrophils; nuclei are stained red
(Fig. 28.10). In chronic myeloid leukemia, LAP is either
absent or low. In leukemoid reaction and in other
myeloproliferative neoplasms (polycythemia vera,
essential thrombocythemia, and myelofibrosis), LAP score
is increased. Therefore, LAP score is useful in
differentiating CML from these disorders. Depending on
the intensity of staining, LAP score is graded in each
neutrophil as follows:
• 0: Negative; No granules
• 1: Positive with very few granules
• 2: Positive with few to moderate number of granules
• 3: Strongly positive with numerous granules
Fig. 28.10: Leukocyte alkaline phosphatase reaction: three
• 4: Very strongly positive with cytoplasm crowded neutrophils with score: 0, two neutrophils with a score of 2,
with granules. and one neutrophil with score of 3
Laboratory Tests in Hematological Malignancies 283

LAP score in an individual smear is the sum of the surface membrane immunoglobulin, single light chain,
scores of 100 consecutive neutrophils. Normal range is CD19, CD20, and T-associated antigen CD5). The
40-100. neoplastic lymphocytes are more fragile than normal cells
Chronic phase of CML should be differentiated from and are often disrupted while spreading blood smear
leukemoid reaction in which myeloid cells increase in producing smudge (or basket) cells (Fig. 28.11).
number in peripheral blood secondary to infections, Complications include infections, autoimmune
inflammation, and other disorders (See Table 22.1 in
hemolytic anemia, and transformation to prolymphocytic
Chapter 22: Blood Smear).
leukemia or large cell lymphoma (Richter’s syndrome).
Accelerated phase: During accelerated phase, basophils
and blast cells increase in number and patient increa- PLASMA CELL DYSCRASIAS
singly becomes resistant to therapy.
Plasma cell dyscrasias are a group of disorders
Blast crisis: Blast crisis results when blast cells become (Table 28.9) characterized by clonal proliferation of
> 20% in blood or bone marrow. Additional cytogenetic plasma cells or plasmacytoid lymphocytes and mono-
abnormalities develop in accelerated and blast phases. clonal production of immunoglobulins.
Features of plasma cell dyscrasias are summarized
Chronic Lymphocytic Leukemia in Table 28.10.
Chronic lymphocytic leukemia (CLL) is a neoplastic Laboratory investigations in plasma cell dyscrasias
disorder characterized by monoclonal proliferation of are shown in Box 28.2.
immunologically incompetent, slowly dividing, mature The laboratory findings that are suspicious of a
B lymphocytes. Most patients are adults >60 years. plasma cell dyscrasia are raised erythrocyte sedimen-
Clinical features include insidious onset of weakness, tation rate, rouleaux formation on blood smear, increased
weight loss, susceptibility to infections, lymphadeno- plasma cells in bone marrow (Fig. 28.12), renal impair-
pathy, and splenomegaly. About 25% of patients are ment with bland urinary sediment, unexplained lytic bone
asymptomatic. lesions, anemia associated with renal failure and bone
Laboratory features include absolute lymphocytosis
(> 5000/cmm) consisting of small mature lymphocytes, Table 28.9: Plasma cell dyscrasias
smudge cells, and typical immunophenotype (weak 1. Multiple myeloma
2. Plasmacytoma
3. Waldenström macroglobulinemia
4. Amyloidosis
5. Heavy chain disease
6. Monoclonal gammopathy of undetermined
significance (MGUS)

Table 28.10: Characteristics of plasma cell dyscrasias

Multiple myeloma: Osteolytic lesions, bone pain, pathological


fractures, raised erythrocyte sedimentation rate, hyper-
calcemia, anemia, renal disease, serum M protein >3 g/dl,
urinary M protein, plasma cells >10% in bone marrow
Solitary plasmacytoma: Single tumor in bone, no urine or
serum protein abnormalities, no myeloma cells in marrow
Waldenström macroglobulinemia: Organomegaly, hyper-
viscocity, Serum IgM >3 g/dl, infiltration of bone marrow with
plasmacytoid lymphocytes
Monoclonal gammopathy of undetermined significance:
No osteolytic lesions/hypercalcemia/anemia/renal disease,
serum M protein <3 g/dl, plasma cells <10% in bone marrow,
Fig. 28.11: Blood smear in chronic lymphocytic leukemia no urinary M protein
showing small mature-looking lymphocytes and a smudge cell
284 Essentials of Clinical Pathology

Box 28.2: Laboratory investigations in


plasma cell dyscrasias

• Serum and urine protein electrophoresis: Screening,


diagnosis, and monitoring the course of disease and
response to therapy
• Immunoelectrophoresis: Diagnosis and identification of
type of M protein
• Serum/urine immunofixation: Diagnosis, identification of
clonal nature by immunotyping
• Quantitation of M protein by radial immunodiffusion:
Diagnosis and follow-up; may help in differentiating benign
from malignant process (e.g. MGUS from multiple
myeloma)
• Serum viscosity: Diagnosis of hyperviscosity syndrome; this
is especially common in Waldenström macroglobulinemia
(due to elevated IgM) and uncommon in IgG-secreting multiple
myeloma. Hyperviscosity symptoms become clinically
apparent when viscosity exceeds 3.0 centipoise.
Fig. 28.12: Bone marrow smear in myeloma showing
plasma cells

Fig. 28.13: Principle of serum protein electrophoresis. (A) On agar gel electrophoresis, serum proteins get separated into
5 regions or bands. Each region or band contains 1 or more proteins, knowledge of which aids in interpretation. (B) Densitometric
scan of serum protein electrophoresis reveals 5 peaks; elevation or depression of the peak(s) can reflect concentrations
of constituent proteins. If abnormal pattern is identified (e.g. M protein), further testing is done to identify the nature of M
protein and quantification of M protein

pain, Bence Jones proteinuria, and hypergamma- on the supporting medium (like agar gel, cellulose,
globulinemia with immune deficiency. etc.) which is then placed in an electrophoresis
1. Serum protein electrophoresis: The most common chamber and exposed to the electric current. Serum
indication for serum protein electrophoresis is proteins get separated into five components: albumin,
suspected plasma cell dyscrasia (especially multiple α1-globulins, α2-globulins, β-globulins, and
myeloma). In this technique, patient’s serum is placed γ-globulins (Fig. 28.13). The last four zones are
Laboratory Tests in Hematological Malignancies 285

Table 28.11: Components of serum protein electrophoresis


Component Proteins Common abnormalities

1. Albumin Albumin ↓ in liver disease, malnutrition,


nephrotic syndrome
2. α1-globulins • α1-antitrypsin ↓ in α1-antitrypsin deficiency
• α1-glycoprotein (orosomucoid)
• Thyroid-binding globulin
3. α2-globulins • α2-macroglobulin ↑ in acute phase response
• Haptoglobin
• Ceruloplasmin
4. β-globulins • Transferrin
• C-reactive protein
• β-lipoprotein
5. γ-globulins Immunoglobulins ↑ severe infection, chronic liver disease,
autoimmune disorders, plasma cell dyscrasias;
↓ hypogammaglobulinemia
↓: Decreased; ↑: Increased

Table 28.12: Comparison of polyclonal and monoclonal gammaglobulinemias


Parameter Polyclonal gammaglobulinemia Monoclonal gammaglobulinemia
1. Pathogenesis Production of immunoglobulins from Production of immunoglobulins from a
single
different clones of plasma cells neoplastic or potentially neoplastic clone of
plasma cells
2. Causes Chronic liver disease, collagen Multiple myeloma, Waldenström
vascular disease, chronic infections, macroglobulinemia, amyloidosis,
metastatic carcinoma monoclonal gammopathy
of undetermined significance, heavy chain
disease
3. Protein electrophoresis A wide band with indistinct borders A localized dense band with sharp borders
4. Densitometer scan A broad-based peak A narrow, tall, sharp spike
5. Nature of light chains Both κ and λ light chains Either κ or λ light chain

composed of a mixture of proteins (Table 28.11). If plasma cell dysrasia is strongly suspected and
Majority of plasma proteins are synthesized in the liver, serum protein electrophoresis is normal, immuno-
except immunoglobulins that are produced by plasma fixation should be performed as it is more sensitive
cells. for detection of small amounts of monoclonal protein.
The bands are inspected visually for any The amount of M protein corresponds directly with
qualitative abnormality. For quantitation, membrane tumour burden.
is then run through an instrument called as densito- Comparison of polyclonal vs. monoclonal
meter. This records absorbance of each protein band, gammaglobulinemia is presented in Table 28.12.
generates a tracing, and quantitates percentage 2. Immunoelectrophoresis: This technique combines
fraction of each band. If total serum proteins have been separation by electrophoresis and identification by
measured earlier, concentration of each protein band double immunodiffusion. Antigenic mixture (placed
can be obtained from the percentage fraction. in a well cut in agar gel) is separated by electro-
286 Essentials of Clinical Pathology

phoresis. A trough is cut parallel to the electrophoretic ponding immunoglobulin protein to form a complex
migration, filled with antibody, and then kept for that becomes fixed in the gel. The gel is washed to
immunodiffusion in a moist chamber for 24 hours. remove the unbound proteins, leaving only the fixed
The resulting immunoprecipitates form arcs that protein that is then visualized with staining (Fig.
assume different shapes and varying mobilities 28.16). The test is sensitive and can detect small
(Figs 28.14 and 28.15). amounts of M protein.
This technique is most useful for recognition of 4. Single radial immunodiffusion (SRID): This is a
abnormal immunoglobulins in paraproteinemias. quantitative method used for measuring concen-
3. Immunofixation electrophoresis: This technique is tration of serum proteins (M protein in parapro-
used on serum or urine to identify the nature of M teinemias). In this technique, antibody is incorporated
protein. In this type of electrophoresis, monospecific into the gel. A soluble antigen is placed in the well
antibodies are used to identify specific types of cut in the agar gel. Following the radial diffusion of
immunoglobulins and light chains. the antigen into the antibody-incorporated gel,
Patient’s serum sample is subjected to agarose gel circular precipitates develop. The diameter of the
electrophoresis in 5 different lanes. Different circular precipitates is directly proportional to the
monospecific antibodies (IgG, IgM, IgA, κ light chain, antigenic concentration, provided that the mono-
λ light chain) are then applied directly over the specific antibody is evenly distributed in the uniformly
surface of gel. The antibody reacts with the corres- thick gel, and the well size and the volume of the

Fig. 28.14: Principle of immunoelectrophoresis


Laboratory Tests in Hematological Malignancies 287

Fig. 28.15: Imunoelectrophoresis. (A) Two wells and central


trough pattern. This is used for comparing immunoelectro-
phoretic pattern of normal serum with that of patient’s serum.
Top well is filled with normal serum while bottom well is filled
with patient’s serum. After electrophoresis, central trough is
charged with monospecific antiserum to IgG. (B) Thickening Fig. 28.17: Principle of single radial immunodiffusion
and bowing of IgG arc from patient’s serum indicates IgG
paraproteinemia. Normal IgG arc is shown for comparison
BIBLIOGRAPHY
1. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for
the classification of the acute leukemias: French-
American-British (FAB) Cooperative Group. Br J
Hematol 1976;33:451-8.
2. Bennett JM, Catovsky D, Daniel MT, et al. Proposal for
the recognition of minimally differentiated acute
myeloid leukemia (AML-M0). Br J Hematol 1991;
78:325-9.
3. Bennett JM, Catovsky D, Daniel MT, et al. Proposed
revised criteria for the classification of acute myeloid
leukemia. Ann Intern Med 1985;103:620-5.
4. Bennett JM, Catovsky D, Daniel MT, et al. The
morphological classification of acute lymphoblastic
leukemia: concordance among observers and clinical
Fig. 28.16: Immunofixation showing monoclonal protein correlations. Br J Hematol 1981;47:553-61.
IgG λ. SPE: Serum protein electrophoresis 5. Jaffe ES, Harris NL, Stein H, Vardiman JW (Eds). World
Health Organization Classification of Tumours.
Pathology and Genetics of Tumours of Hematopoietic
antigen are kept constant. The concentration can be and Lymphoid Tissues. Lyon; IARC Press, 2001.
read from the reference graph prepared earlier using a 6. Kawthalkar SM. Essentials of Hematology. New Delhi:
set of standards (Fig. 28.17). Jaypee Brothers and Medical Publishers (P) Ltd, 2006.
29
Laboratory Tests in
Bleeding Disorders

PHYSIOLOGY OF HEMOSTASIS cells synthesize von Willebrand factor (vWF), tissue factor,
and platelet activating factor, which promote hemostasis.
Hemostasis is the normal physiologic mechanism for
Also, following injury, subendothelial collagen is exposed
keeping the blood in fluid state in vascular system and for
which provides site for attachment of platelets (adhesion).
prevention of hemorrhage by complex interaction of blood
Endothelial cells synthesize prostacycline (inhibits platelet
vessel walls, platelets, and plasma proteins. Following
aggregation), protein S (a cofactor for protein C, which is
injury, initially vessel wall and platelets interact to control
an inhibitor of coagulation), and tissue plasminogen
hemorrhage by forming a platelet plug at the site of injury;
activator (activates fibrinolysis).
this is called as primary hemostasis. This is followed by
activation of coagulation factors by a series of enzymatic
Platelets
reactions to form a stable fibrin clot (platelet plug
enmeshed by fibrin); this is secondary hemostasis. Platelets are produced by cytoplasmic fragmentation of
Dissolution of the clot will eventually occur by fibrinolysis megakaryocytes in bone marrow. Life-span of platelets is
when healing is complete. Hemostatic equilibrium about 7-10 days. Normal platelet count in peripheral blood
requires normal blood vessels, normal platelets, normal is 1.5-4.0 lac/µl. About 2/3rd of platelets in the body are
coagulation factors, normal fibrinolysis, and normal circulating in peripheral blood, while 1/3rd are pooled in
coagulation inhibitor system.
spleen.
Role of the three main components of hemostasis is
Main functions of platelets in hemostasis are adhesion,
outlined below.
release reaction, and aggregation (Fig. 29.1).
Blood Vessel Wall Platelets attach to exposed subendothelium following
Transient constriction of blood vessels occurs at the site injury; this adhesion is mediated by vWF, which binds
of injury which helps to control blood loss. Endothelial to GpIb receptor on platelets. This initiates activation of

Fig. 29.1: Platelet adhesion and aggregation


Laboratory Tests in Bleeding Disorders 289

platelets, which change shape from a disc to a sphere and Coagulation System
release their contents to the exterior (release reaction). Normally, coagulation factors (Table 29.1) are circulating
These are mainly adenosine diphosphate (ADP), in an inactive form. Except for thromboplastin and
serotonin, and thromboxane A 2 (TxA 2 ). Platelet calcium, all the coagulation factors are proteins.
aggregation refers to sticking of platelets to each other; Coagulation factors have been divided into thee groups
platelet agonists such as collagen, ADP, thrombin, and depending on similarities in structural and functional
TxA2 mediate aggregation. Binding of ADP to platelets properties: (1) Fibrinogen group: I, V, VIII, XIII; (2)
causes exposure of GpIIb/IIIa receptors which bind Vitamin-K dependent: II, VII, IX, X; and (3) Contact
fibrinogen. Fibrinogen binding to multiple platelets group: XI, XII, high molecular weight kininogen,
causes formation of large platelet aggregates. prekallikrein. When activated, coagulation factors
Activated platelets also provide phospholipid surface interact with each other in a sequential manner to
for certain coagulation reactions (platelet factor 3 or ultimately form a fibrin clot and arrest bleeding. Blood
platelet procoagulant activity). coagulation occurring in vitro is divided into three
pathways: extrinsic, intrinsic, and common (Fig. 29.2).
Plasma Proteins This division is helpful in understanding the principles
Plasma proteins, which regulate hemostasis, are of common screening tests of coagulation.
coagulation factors, coagulation inhibitors, and proteins Extrinsic pathway: The extrinsic pathway is initiated when
of fibrinolytic system. F VII combines with tissue factor (released after tissue
Table 29.1: Coagulation factors

Factor Synonym Site of synthesis Significant features

I Fibrinogen Liver Normal: 200-400 mg/dl; absent in serum


II Prothrombin Liver Vitamin K-dependent; multiple actions; absent in
adsorbed plasma
III Tissue factor; Various tissues Activates coagulation through
thromboplastin extrinsic pathway
IV Calcium Obtained from Acts as a cofactor in various
diet and bones coagulation reactions
V Labile factor Liver, platelets Cofactor in conversion of
prothrombin to thrombin; absent in aged plasma
VI There is no factor VI
VII Stable factor Liver Vitamin K-dependent; absent in
adsorbed plasma; sensitive to
oral anticoagulant therapy
VIII Antihemophilic globulin; Liver Two subunits: VIII:C and VIII:vWF; cofactor in the
antihemophilic factor conversion of F X to F Xa; absent in aged plasma
IX Christmas factor Liver Vitamin-K dependent; absent in adsorbed plasma
X Stuart-Prower factor Liver Vitamin-K dependent; absent in adsorbed plasma
XI Plasma thromboplastin Liver Contact factor
antecedent
XII Hageman factor; Liver Contact factor; activates coagulation in vitro;
Contact factor deficiency does not cause bleeding
XIII Fibrin stabilizing factor; Liver, platelets Stabilizes fibrin clot by cross-linking fibrin
Laki-Lorand factor monomers
Prekallik- Fletcher factor Liver Contact factor; has many functions
rein
High Fitzgerald factor Liver Contact factor; has many functions
molecular
weight
kininogen

Aged plasma: Stored plasma that is deficienct in factors V and VIII. Adsorbed plasma: Plasma adsorbed with ammo-
nium hydroxide that is deficient in factors II, VII, IX, and X (vitamin K-dependent factors)
290 Essentials of Clinical Pathology

Fig. 29.2: Normal coagulation mechanism. HMWK = high molecular weight kininogen; PL = phospholipid; Ca = calcium;
TF: tissue factor; solid arrow: conversion; Dotted arrow: action; subscript a: activated form of the coagulation factor

injury) in the presence of calcium ions. F VII-tissue factor Inhibitors of Coagulation


complex in turn activates F X to F Xa.
Several physiologic inhibitors of coagulation are present
Intrinsic pathway: Intrinsic system is initiated when F XII in circulation to ensure that the fibrin clot remains
is activated in vitro ( e.g. by glass). This activation requires localized to the site of injury and activation of coagulation
presence of high molecular weight kininogen (HMWK) is short-lived. Important inhibitors of coagulation are
and prekallikrein. The roles of F XII, HMWK, and tissue factor pathway inhibitor (released from blood
prekallikrein in coagulation in vivo are not clear since vessels; neutralizes VII-tissue factor complex), anti-
their deficiencies are not associated with bleeding. thrombin III (most important physiological inhibitor;
Activated F XII activates F XI, which in turn activates F synthesized in liver; heparin-like substances on
IX to F IXa. F IXa combines with F VIII, phospholipid, endothelial cells promote activity; inhibits mainly
and calcium to activate F X to F Xa. thrombin and F Xa), and protein C (activated by thrombin
Common pathway: Activation of F X to F Xa, by either in the presence of thrombomodulin on the surface of
extrinsic or intrinsic mechanism, marks the beginning endothelial cells; causes proteolysis of activated forms
of common pathway. F Xa binds FV, phospholipid, and of F V and F VIII; protein S acts as a cofactor in this
calcium and converts prothrombin to thrombin. reaction) (Fig. 29.3).
Thrombin splits off fibrinopeptides A and B from
Fibrinolytic System
fibrinogen to form fibrin monomers, which sponta-
neously polymerize. F XIII stabilizes fibrin polymers by Fibrinolysis is the process of dissolution of blood clot to
introducing covalent bonds between adjacent poly- ensure free flow of blood in vascular system. During
peptide chains. coagulation process, plasminogen binds to fibrin
Laboratory Tests in Bleeding Disorders 291

Fig. 29.3: Natural inhibitors of coagulation and their actions. – indicates inhibition. Antithrombin III mainly inhibits F Xa and
thrombin, activated protein C degrades activated forms of FV and F VIII, while tissue factor pathway inhibitor inhibits F VII-
tissue factor complex

Fig. 29.4: Mechanism of fibrinolysis

network. Endothelial cells secrete tissue plasminogen Fibrinolysis is inhibited by plasminogen activator
activator, which activates plasminogen to plasmin. inhibitor (PAI) type 1, α2-antiplasmin and α2-macro-
Plasmin cleaves fibrin strands to release fibrin globulin.
degradation products. Plasmin also digests fibrinogen BLEEDING DISORDERS
(Fig. 29.4). Fibrinogen/fibrin degradation products Bleeding disorders are the result of a generalized defect in
interfere with action of thrombin and with platelet hemostasis due to abnormalities of blood vessels, platelets,
aggregation. FDPs are cleared from the circulation by or coagulation factors. Common bleeding disorders are
macrophages. listed in Table 29.2.
292 Essentials of Clinical Pathology

Table 29.2: Bleeding disorders

Disorders of blood vessels


Acquired Hereditary
Allergic purpura Hereditary hemorrhagic telangiectasia
Infections: Gram-negative septicemia, viral infections
Scurvy
Senile purpura
Disorders of platelets
Thrombocytopenia Defective platelet function
Increased destruction of platelets Acquired
Idiopathic thrombocytopenic purpura Drugs: aspirin, other nonsteroidal anti-inflammatory drugs,
antibiotics, xylocaine
Infections: malaria, dengue, septicemia, subacute Myeloproliferative disorders
bacterial endocarditis, rubella, infectious mononucleosis
Drugs: quinidine, quinine, heparin, procainamide Uremia
Heparin induced thrombocytopenia Paraproteinemia
Disseminated intravascular coagulation Hereditary
Massive blood transfusion Storage pool deficiency
Thrombotic thrombocytopenic purpura Bernard-Soulier syndrome
Alcohol Glanzmann’s thrombasthenia
Toxemia of pregnancy
Increased pooling of platelets in spleen
Hypersplenism
Decreased production of platelets in bone marrow
Aplastic anemia
Bone marrow infiltration: leukemias, lymphomas, myeloma
Megaloblastic anemia
Drugs
Infections
Radiation
Hereditary disorders
Disorders of coagulation
Hereditary Acquired
Hemophilia A (F VIII deficiency) Disseminated intravascular coagulation
von Willebrand’s disease Liver disease
Hemophilia B (F IX deficiency) Vitamin K deficiency
Disorders of fibrinogen Massive blood transfusion
Heparin or oral anticoagulant therapy
Renal disease
Paraproteinemia
Inhibitors of coagulation

Disorders of Blood Vessels present if platelet count is < 1,50,000/cmm. Causes of


Vascular disorders can result in easy bruising and muco- thrombocytopenia are listed in Table 29.2. Bleeding
cutaneous type of bleeding. Vascular disorders may be manifestations include petechie, purpura, ecchymoses,
inherited or acquired (Table 29.2) and defect may be in and mucous membrane bleeding.
the connective tissue lining (collagen or elastic tissue) or
in endothelial cells. Immune Thrombocytopenic Purpura (ITP)
Disorders of Platelets ITP is of two types: acute and chronic. Acute ITP has a
Disorders of platelets can be divided into disorders of sudden onset, occurs usually in children, and often
platelet number or of function. Thrombocytopenia is follows a viral infection. Platelet count is very low.
Laboratory Tests in Bleeding Disorders 293

Complete remission is usual. Chronic ITP has a gradual seen in children. It is characterized by a triad of acute
onset, occurs usually in adults (especially young females), renal failure, thrombocytopenia, and microangiopathic
and platelet count is moderately decreased. Cause is hemoloytic anemia.
unknown. It is characterized by remissions and
exacerbations over a long duration. Bone marrow Post-Transfusion Purpura
examination in ITP shows increased number of mega- This is allo-antibody induced thrombocytopenia. There is
karyocytes. Diagnosis of ITP requires exclusion of all a sudden onset of severe thrombocytopenia in some adult
other causes of thrombocytopenia. maltiparous women 1-10 days following blood
transfusion. It is due to sensitization to platelet antigen
Thrombotic Thrombocytopenic Purpura HPA-1a (platelet antigen A1 or PlA1) during previous
pregnancy by foetal platelets.
There is formation of hyaline microthrombi in micro-
circulation due to systemic clumping of platelets because Disorders of Platelet Function
of unusually large multimers of von Willebrand factor.
This causes thrombocytopenia. The characteristic pentad Disorders of platelet function are characterized by
of signs includes: bleeding secondary to severe thrombo- prolonged bleeding time and normal platelet count. They
cytopenia, microangiopathic hemolytic anemia (with may be inherited or acquired. Acquired disorders are
production of schistocytes), fever, neurological signs, and more common. Sites of defects in platelet function
renal abnormalities. disorders are shown in Figure 29.5.

Bernard Soulier syndrome: This is a rare, autosomal


Hemolytic Uremic Syndrome
recessive disorder with severe bleeding manifestations.
It is caused by ingestion of food contaminated with There is a congenital absence of platelet membrane
verotoxin-producing Escherichia coli and is commonly glycoprotein complex GpIb/IX leading to failure of

Fig. 29.5: Sites of defects in disorders of platelet function


294 Essentials of Clinical Pathology

adhesion of platelets to subendothelium via von gene. It is an X-linked recessive disorder primarily affecting
Willebrand factor. Blood smear shows giant platelets. males; females are carriers but do not manifest the disease
Platelet aggregation studies reveal impaired aggregation (Fig. 29.6). Hemophilia A is classified into three types
with ristocetin and normal aggregation with other based on the level of F VIII level in plasma: mild, moderate,
agonists. and severe (Table 29.4).
Glanzmann’s thrombasthenia: In this very rare autosomal In severe hemophilia, hemarthroses lead to crippling
recessive disorder, platelet aggregation is defective due deformities; intramuscular hematomas can compress
to congenital absence of platelet membrane glycoproteins vital structures; intracranial hemorrhage can occur
GpIIb/IIIa on platelet surface. Laboratory features following minor trauma; operative and post-traumatic
include discrete, small platelets on blood smear, poor clot bleeding can be life-threatening; infections like hepatitis
retraction, and defective platelet aggregation with ADP, and acquired immunodeficiency syndrome can be
epinephrine, and collagen, and normal aggregation with transmitted through blood products. Screening tests for
ristocetin. hemostasis show normal bleeding time, platelet count,
and prothrombin time. Activated partial thromboplastin
Aspirin inhibits the enzyme cyclo-oxygenase leading to
time is prolonged. Diagnosis is made by one-stage F VIII
failure of synthesis of thromboxane A2 that is required
assay.
for platelet aggregation. The inhibitory effect of aspirin
on platelets lasts for 7-10 days (i.e. lifespan of platelets).
Hemophilia B (Christmas Disease, F IX Deficiency)
Inherited Disorders of Coagulation
This is clinically indistinguishable from hemophilia A.
Deficiencies of all the coagulation factors have been
reported. Out of these, the three relatively common Diagnosis requires F IX assay.
disorders are hemophilia A (F VIII deficiency), hemo-
philia B (F IX deficiency), and von Willebrand disease von Willebrand Disease (VWD)
(Table 29.3).
vWD is a markedly heterogeneous congenital bleeding
Hemophilia A disorder characterized by deficiency or functional defect
(Classical Hemophilia, F VIII Deficiency) of von Willebrand factor (vWF). Mode of inheritance is
It is caused by hereditary deficiency or dysfunction of F autosomal dominant or recessive, with overall prevalence
VIII due mainly to point mutations or deletions of F VIII in the general population being 1%. There are three main

Table 29.3: Inheritance and incidence of inherited bleeding disorders


Disorder Inheritance Incidence
von Willebrand Autosomal dominant 1:100
disease or recessive
Factor VIII deficiency X-linked recessive 1:10000
Factor IX deficiency X-linked recessive 1:60000
Factor VII deficiency Autosomal recessive 1:500,000
Fibrinogen deficiency Autosomal recessive 1:1 million
Factor V deficiency Autosomal recessive 1:1 million
Factor X deficiency Autosomal recessive 1:1 million
Factor XI deficiency Autosomal recessive 1:1 million
Factor XIII deficiency Autosomal recessive 1:2 million
Prothrombin deficiency Autosomal recessive 1:2 million
Note: Deficiencies of F XII, high molecular weight kininogen, and prekallikrein are not associated with bleeding
Laboratory Tests in Bleeding Disorders 295

Fig. 29.6: Typical pedigree in X-linked recessive trait (hemophilia A or B). Females are
heterozygous (carriers) and only males are affected

Table 29.4: Classification of hemophilia


Type F VIII level Clinical features
Mild >5% Excess bleeding only after major trauma or surgery
Moderate 1-5% Excess bleeding after mild to moderate trauma; occasional
hemarthrosis; spontaneous bleeding infrequent
Severe <1% Frequent and spontaneous deep tissue hematomas and
hemarthroses

Table 29.5: Comparison of three main inherited bleeding disorders


Feature Von Willebrand disease Hemophilia A Hemophilia B
1. Incidence 1:100 1:10,000 1:60,000
2. Inheritance AD or AR XR XR
3. Sex affected Male or female Male Male
4. Nature of bleeding Superficial (Mucocutaneous) Deep (muscles, joints) Deep (muscles, joints)
5. Screening tests
• BT Increased Normal Normal
• PC Normal or low Normal Normal
• PT Normal Normal Normal
• APTT Normal or increased Increased Increased
6. Specific test Platelet aggregation with F VIII assay F IX assay
for diagnosis ristocetin

AD: Autosomal dominant; AR: Autosomal recessive; XR: X-linked recessive; BT: Bleeding time; PC: Platelet count;
PT: Prothrombin time; APTT: Activated partial thromboplastin time

types: I, II, and III. Type I (partial deficiency of vWF) is the Screening tests for hemostasis reveal prolonged bleeding
most common in which all types of vWF multimers (small, time and activated partial thromboplastin time. Platelet
intermediate, and large) are mildly deficient; bleeding count and prothrombin time are normal. Ristocetin-
manifestations are slight. In type II vWD (qualitative
induced platelet aggregation is deficient. One stage F VIII
defects in vWF) there is a qualitative abnormality of vWF
assay shows reduced F VIII activity.
with absence of large vWF multimers. Type III (complete
vWF deficiency) is a rare severe bleeding disorder in which The three main inherited bleeding disorders are
there is a severe deficiency of all forms of vWF multimers. compared in Table 29.5.
296 Essentials of Clinical Pathology

Acquired Disorders of Coagulation

Disseminated Intravascular Coagulation (DIC)


This is an acquired thrombohemorrhagic disorder
occurring as a secondary complication in a wide
spectrum of disorders, and characterized by (i) activation
of coagulation with formation of microthrombi in
microcirculation, (ii) activation of fibrinolysis, and (iii)
consumption of platelets, coagulation factors, and fibrin
leading to bleeding diathesis.
Important causes of DIC include sepsis or severe
infections, massive crush injury, obstetric complications
(amniotic fluid embolism, placental abruption, intra-
uterine retention of dead fetus, eclampsia, septic
abortion), malignancy (disseminated mucin-secreting
adenocarcinoma, acute promyelocytic leukemia), snake
bite, shock, heat stroke, severe liver disease, and acute
intravascular hemolysis. DIC can be initiated due to Fig. 29.7: Blood smear in acute disseminated
above causes by: (i) release of tissue factor or thrombo- intravascular coagulation showing fragmented red cells
plastic substances in circulation, or (ii) endothelial cell
damage.
newborn, and in adults with poor dietary intake,
There are two main presentations: acute and chronic.
malabsorption , obstructive jaundice, and drugs like oral
Acute or uncompensated DIC is a severe disease with
anticoagulants.
significant bleeding manifestations. Chronic DIC is a
mild, protracted disease characterized mainly by venous Hemorrhagic disease of newborn Normally, vitamin K-
thrombosis. Laboratory features in DIC are shown in Box dependent factors are low at birth; vitamin K deficiency
29.1. exaggerates this deficit and causes bleeding. In classic
cases, hemorrhage manifests around 2-4 days of life.
Screening tests reveal normal platelet count and
Box 29.1: Laboratory features in disseminated
intravascular coagulation prolongation of both prothrombin time and activated
partial thromboplastin time.
• Acute DIC: Platelet count: Low, Prothrombin time:
Prolonged, Activated partial thromboplastin time: Pro-
Liver Disease
longed, Blood smear: Fragmented red cells (Fig. 29.7),
Fibrinogen level: Low, Fibrinogen/fibrin degradation Pathophysiology of hemostatic defect in liver disease is
products: Increased, D-dimer: Increased
complex: (i) deficient synthesis of coagulation factors in
• Chronic DIC: Platelet count, Prothrombin time, Activated hepatocellular disease, (ii) deficient synthesis of vitamin
partial thromboplastin time: Normal; Fibrinogen/fibrin
degradation products, D-dimer: Increased. K-dependent factors in biliary obstruction, (iii) synthesis
of dysfunctional fibrinogen (dysfibrinogenemia), (iv)
decreased clearance of activated coagulation factors, (v)
Vitamin K Deficiency defective platelet function due to raised fibrinogen/fibrin
Vitamin K is a fat-soluble vitamin necessary for the degradation products, or (vi) disseminated intravascular
synthesis of coagulation factors II, VII, IX, and X, and also coagulation.
two natural anticoagulant proteins C and S. Vitamin K is
required for gamma carboxylation of glutamic acid Acquired Inhibitors of Coagulation
residues of the above coagulation factors. Vitamin K, (Circulating Anticoagulants)
being fat-soluble, requires bile salts for absorption. Acquired inhibitors of coagulation are of two main types:
Vitamin K deficiency occurs in hemorrhagic disease of specific and non-specific. Most common specific inhibitor
Laboratory Tests in Bleeding Disorders 297

is an antibody directed against F VIII. It develops in multi- Clinical Evaluation


transfused patients of F VIII deficiency. Inhibitors render
Significant bleeding may result from a local cause or a
F VIII replacement therapy ineffective. Autoantibodies
bleeding disorder (generalized hemostatic defect). A
against F VIII can also develop in old age, rheumatoid
bleeding disorder is suggested by the presence of easy
arthritis, and postpartum females.
bruising, spontaneous bleeding, bleeding from multiple
Non-specific inhibitors are lupus inhibitors that
sites, repeated episodes of excessive bleeding, restart of
develop in some patients with systemic lupus erythe-
bleeding hours or days following injury, similar past or
matosus, other autoimmune disorders, neoplasia, and
family history, history of poor wound healing or when
human immunodeficiency virus infection. These are
bleeding is out of proportion to the degree of trauma.
antiphospholipid antibodies which interfere with
coagulation reactions that need phospholipid surface. Nature of bleeding can help in differentiating
Patients usually have recurrent venous thromboses, between primary and secondary disorders of hemostasis:
repeated spontaneous abortions, and intrauterine fetal • Petechia: Tiny pinpoint areas of red to purple
death. hemorhage (< 2 mm in diameter) on skin or mucous
Coagulation inhibitors cause prolongation of APTT membranes due to a primary hemostatic disorder.
and are detected by mixing experiment (see later). F VIII • Purpura: Areas of hemorrhage in skin or mucous
inhibitor is measured by Bethesda inhibitor assay. For membrane, red to purple in color, non-blanching and
detection of lupus anticoagulant, various tests are >3 mm but <1 cm in diameter. They occur in disorders
available like kaolin clotting time, dilute Russell’s viper of primary hemostasis.
venom time, and platelet neutralization assay. • Ecchymosis (Bruise): An area of extravasated blood in
skin > 1 cm in diameter. It may be due to defective
APPROACH TO THE DIAGNOSIS OF hemostasis or trauma.
BLEEDING DISORDERS • Hematoma: A swelling resulting from a large area of
hemorrhage in subcutaneous tissue or muscle. It
A systematic approach, consisting of both clinical and occurs in coagulation disorders.
laboratory evaluation, is necessary for diagnosis of • Hemarthrosis: This refers to bleeding into a joint. It
bleeding disorders (Fig. 29.8). results from a coagulation disorder, especially
hemophilia.
Bleeding manifestations typical of some bleeding
disorders are:
• Muco-cutaneous bledding or bleeding from skin and
mucous membranes (purpura, petechia, bleeding from
gums, epistaxis, gastrointestinal/rectal bleeding,
menorrhagia): Platelet disorders, von Willebrand
disease.
• Cephalhematomas, hemarthrosis, intramuscular
hematoma, intracerebral bleeding, retroperitoneal
hemorrhage: hemophilia, von Willebrand disease,
afibrinogenemia
• Umbilical stump bleeding: afibrinogenemia, F XIII
deficiency
• Defective wound healing: F XIII deficiency
• Recurrent severe epistaxis: hereditary hemorrhagic
telangiectasia
Detailed family history should be elicited and a family
tree constructed. History of similar abnormality in close
relatives is suggestive of a hereditary disorder. From the
Fig. 29.8: General approach for investigation of a pattern of inheritance, diagnostic possibilities can be
bleeding disorder narrowed and investigations can be directed accordingly.
298 Essentials of Clinical Pathology

History of bleeding only in males and positive family Laboratory Evaluation


history on maternal side spanning many generations is Initial tests, which should be performed in a suspected
suggestive of an X-linked disorder (e.g. F VIII or F IX bleeding disorder, are complete blood count including
deficiency). In autosomal recessive diseases (e.g. blood smear, platelet count, bleeding time, clotting time,
afibrinogenemia, deficiency of FV or FX, von Willebrand prothrombin time, and activated partial thromboplastin
disease), both males and females are affected (in the same time. Depending on the results of these screening tests,
generation) and history of consanguinity in parents is one or more specific tests are carried out for definitive
common. In autosomal dominant disorders (e.g. some diagnosis (e.g. platelet function studies, assays of
types of von Willebrand disease), bleeding manifests in coagulation factors, and test for fibrin degradation
both sexes, in one parent and also in older generations. products) (Table 29.7). Abnormalities of blood vessels
Various acquired conditions can cause a generalized are usually not detectable by laboratory tests for
hemostatic defect. These include diseases of liver, renal hemostasis, and their diagnosis requires correlation of
diseases, autoimmune disorders, malabsorption, clinical and other investigations.
malignancy, infections, and administration of drugs (e.g.
heparin, oral anticoagulants, aspirin and other nonsteroi- Screening Tests for Hemostasis
dal anti-inflammatory drugs). Important features to look
Complete Blood Count including Blood Smear
for on physical examination include fever, splenomegaly,
hepatomegaly, lymphadenopathy, and icterus. A complete blood count and a blood smear can provide
A bleeding disorder can be placed into the category information in the form of:
of either vascular/platelet or coagulation abnormality • Presence of cytopenia (anemia, leukopenia,
on the basis of clinical features (Table 29.6). thrombocytopenia)

Table 29.6: Differences between coagulation and platelet/vascular disorders


Parameter Coagulation disorder Platelet/vascular disorder
1. Sex More common in males More common in females
2. Family history Often positive Often negative
3. Petechie, bleeding gums, Rare Common
nose bleeds, melena
4. Skin hemorrhages Large and few Small and many
5. Deep hematomas (muscle Common Not seen
bleeding), hemarthrosis
(joint bleeding)
6. Delayed bleeding Characteristic (12-24 hours Not seen
(recurrence of bleeding hours after injury)
or days following injury)

Table 29.7: Laboratory evaluation of bleeding disorders


Screening tests Specific tests

• Complete blood count and blood smear • Platelet aggregation studies


• Platelet count • Coagulation factor assays
• Bleeding time • Estimation of fibrinogen
• Platelet function analyzer-100 • Test for fibrinogen/fibrin degradation products
• Clotting time • Test for D-dimer
• Prothrombin time • Platelet glycoprotein analysis
• Activated partial thromboplastin time
• Thrombin time
Laboratory Tests in Bleeding Disorders 299

• Red cell abnormalities (especially fragmented red 2. Charge the improved Neubauer counting chamber as
cells which may indicate disseminated intra- described for total leukocyte count.
vascular coagulation) 3. Place the mounted counting chamber inside a moist
• White cell abnormalities (like abnormal cells in chamber (which consists of a covered Petri dish with
leukemias) a dampened filter paper at the bottom) and leave it
• Abnormalities of platelets: thrombocytopenia undisturbed for 20 minutes. This allows platelets to
(normally there is 1 platelets per 500-1000 red settle and prevents drying of fluid.
cells), giant platelets (seen in myeloproliferative
4. Place the charged counting chamber on the stage of
disorders and Bernard-Soulier syndrome), and
the microscope. With the illumination reduced to give
isolated discrete platelets without clumping in
sufficient contrast, bring the central large square
finger-prick smear (seen in uremia, Glanzmann’s
under the focus of the low power (×10) objective.
thrombasthenia).
Changing to ×40 objectives, count the total number
of platelets in five smaller squares (Fig. 29.9). Platelets
Platelet Count
appear as bluish, round or oval, small, brightly
Platelets can be counted manually under a microscope refractile fragments with one or more fine dendritic
or by means of an automated hematology analyzer. processes.
Platelets are difficult to count by manual method since
5. Calculation:
they are small in size (2-4 μm) and difficult to distinguish
from dirt particles and cell debris. Platelet count/μl of blood =
Number of platelets counted × Correction for dilution × Correction
Manual Method for volume
Platelet count/L of blood = Number of platelets counted × 109
Principle: Whole blood sample is mixed with a diluent
(1% ammonium oxalate) in which red cells are lysed. An Note:
improved Neubauer counting chamber is filled with the 1. For counting platelets, phase-contrast microscope is
mixture, and platelets are counted under the microscope. preferable to ordinary light microscope. This is
Result is expressed as number platelets per μl or per liter. because platelets are easily distinguished from dirt
Equipment: Similar to that required for total leukocyte
count (see Chapter 20: Total Leukocyte Count).
Reagent: This is 1% ammonium oxalate. It is prepared by
dissolving 1.0 gm of ammonium oxalate in 100 ml of
distilled water. It should be stored in a refrigerator. Since
small particles can be mistaken for platelets, reagent
should be filtered immediately before use.
Specimen: Venous blood anticoagulated with EDTA is
preferable. Capillary blood should be avoided since
platelets adhere to the skin puncture site resulting in a
erroneously lower platelet count. Platelet count from skin
puncture blood is also not reproducible.

Method
1. Take 0.38 ml of 1% ammonium oxalate in a test tube.
To this add 20 µl of well-mixed anticoagulated
venous blood and mix thoroughly. Dilution of blood Fig. 29.9: Area (P) for counting platelets in
is 1:20. Neubauer chamber
300 Essentials of Clinical Pathology

particles or debris using phase-contrast. A flat-bottom, Clinical significance: Platelet count is usually obtained if
thin, phase-contrast hemocytometer with Neubauer there is a suspicion of a bleeding disorder. Thrombo-
ruling is preferable. cytopenia between 1,50,000-50,000/μl is generally not
2. Platelet count done on blood obtained by skin associated with bleeding. Platelet count between 50,000-
puncture is significantly lower due to adhesion of 20,000/μl causes excess bleeding following surgery or
platelets to the puncture site. Therefore, anticoagu- mild degree of spontaneous bleeding. Platelet count
below 20,000/μl is usually associated with spontaneous,
lated venous blood should be used (If capillary blood
severe hemorrhage. Bleeding is often serious if platelets
is used, a free flowing blood drop should be obtained
are below 5,000/μl. Thrombocytosis (platelet count
after wiping away the first drop of blood). > 4,00,000/μl) in chronic myeloproliferative disorders is
3. Blood and the anticoagulant should be mixed sometimes associated with thrombosis and bleeding
thoroughly by inverting the tube for 20 times. Venous manifestations.
blood sample anticoagulated with EDTA should not Causes of thrombocytopenia: Thrombocytopenia is defined
contain any clots. as platelet count below 1,50,000/μl. Causes of thrombo-
4. All the glassware must be scrupulously clean and the cytopenia are listed earlier in Table 29.2. Evaluation of
diluent fluid must be filtered just before use. This is thrombocytopenia is shown in Figure 29.10.
to prevent erroneous counting of dirt particles as Causes of thrombocytosis: Thrombocytosis is defined as
platelets. platelet count greater than 4,00,000/μl. Its causes are (i)
5. A well-spread blood smear should always be primary: chronic myeloproliferative disorders like
examined simultaneously to check the direct platelet chronic myeloid leukemia, essential thrombocythemia,
count on hemocytometer. Normally there is one idiopathic myelofibrosis, and polycythemia vera; (ii)
platelet per 500-1000 red cells on a blood smear. A secondary (reactive): disseminated malignancy,
rough estimate about number of platelets (adequate, hemorrhage, splenectomy, chronic inflammation, and
low, or increased) thus can be obtained. iron deficiency anemia with bleeding.

Fig. 29.10: Evaluation of thrombocytopenia


Laboratory Tests in Bleeding Disorders 301

Automated Method pressure (40 mm Hg). A disadvantage with this method is


closure of puncture wound before cessation of bleeding.
Automated hematology analyzers more precisely count
Template method uses a special surgical blade, which
platelets. However, these are expensive and have high
running costs. Platelet counting is usually based on the makes a larger cut (5 mm long and 1 mm deep). Although
template method is better, it can produce a large scar and
principle of aperture impedance. A smaller aperture tube
even a keloid in predisposed individuals. Ivy’s method is
is required for platelets and a threshold level needs to be
described below.
set to exclude larger red cells and smaller debris. False
elevation of platelet count by this method can result from Ivy’s method
the presence of fragments of red or white cells, Principle: Three standard punctures are made with a
microspherocytes, and elevated cryoglobulins. Causes of lancet on the volar surface of the forearm under standard
pseudothrombocytopenia (falsely low platelet count) are: pressure, and the average time required for bleeding to
clumping of platelets in EDTA-anticoagulated venous cease from the puncture sites is measured.
blood sample (due to the presence of EDTA-dependent Equipment
platelet antibody in some patients which is active only 1. Sphygmomanometer
in vitro), platelet satellitism (adherence of platelets to 2. Sterile disposable lancets (2-2.5 mm blade with
neutrophils in EDTA-anticoagulated sample), platelet shoulder, which limits the depth of penetration).
clumping due to the presence of platelet cold agglutinins 3. Stopwatch
in blood, and presence of giant platelets (which are not 4. Filter paper
counted as platelets by electronic cell analyzers).
Some analyzers can measure platelet distribution width Method
(PDW), which is a measure of degree of variation of 1. A sphygmomanometer cuff is wrapped around the
platelet size present in a blood sample. High PDW is seen upper arm and inflated to 40 mm of Hg.
2. The dorsal surface of the forearm is cleansed with
in myeloproliferative disorders. In secondary thrombo-
70% ethanol and allowed to dry.
cytosis, PDW is normal.
3. Three punctures are made (about 5 cm apart) in quick
Mean platelet volume (MPV) is increased when
succession with a lancet (Superficial veins, and scars
thrombocytopenia is due to peripheral platelet
or bruises should be avoided).
destruction and is normal or low when thrombo-
4. A stopwatch is started as soon as a puncture is made.
cytopenia is due to defective platelet production.
One stopwatch is needed for each puncture.
Young platelets with residual RNA are called as
5. Blood oozing from the puncture wound is gently
reticulated platelets. These can be identified by some
blotted with a filter paper at 15 seconds intervals.
analyzers and flow cytometers. Increased numbers are
Avoid directly touching the edges of the wound.
seen in idiopathic thrombocytopenic purpura and lower 6. The timer is stopped when blood no longer stains the
numbers in aplastic anemia. filter paper.
7. Time required for bleeding to cease from all the three
Bleeding Time puncture wounds is noted. The average time is
reported as the bleeding time.
The bleeding time test assesses primary hemostasis
8. Sterile adhesive strip is applied over the puncture.
(vascular and platelet components) and is dependent on
adequate functioning of platelets and blood vessels. Reference range: 2-7 minutes. Majority of individuals have
In this test, a superficial skin puncture or incision is bleeding time less than 4 minutes. It should be reported
made and the time required for bleeding to stop is in minutes or nearest half minute. If bleeding continues
measured. Three methods are commonly used: Duke’s, beyond 20 minutes, BT should be reported as >20 minutes
Ivy’s, and template. Duke’s method, which measures and the test is discontinued.
bleeding time following ear lobe puncture, is not Causes of prolongation of bleeding time
advocated since it cannot be standardized and can cause 1. Thrombocytopenia: If platelet count is less than
a large local hematoma. In Ivy’s method, 3 punctures 1,00,000/ml, bleeding time should not be performed,
are made on the volar surface of the forearm with a lancet as it will be prolonged. With a very low platelet count,
(cutting depth 2-2.5 mm) under standardized venous bleeding may be difficult to control.
302 Essentials of Clinical Pathology

Fig. 29.11: Evaluation of prolonged bleeding time

2. Disorders of platelet function time is prolonged. Also, in the presence of a strong clinical
3. von Willebrand disease suspicion of a platelet function defect and normal PFA-
4. Disorders of blood vessels 100 result, further testing is still necessary.
Evaluation of prolonged bleeding time is shown in
Clotting Time
Figure 29.11.
This is a crude test and is now replaced by activated
Platelet Function Analyzer (PFA-100) partial thromboplastin time. Clotting time measures the
This is a newly introduced screening test for platelet time required for the blood to clot in a glass test tube
function that assesses both platelet adhesion and kept at 37°C. Prolongation of clotting time only occurs
aggregation. This method uses an instrument called as in severe deficiency of a clotting factor and is normal in
PFA-100 in which anticoagulated whole blood is passed mild or moderate deficiency.
at a high shear rate through small membranes that have
Prothrombin Time (PT)
been coated with either collagen and epinephrine or
collagen and ADP. Platelets adhere to each membrane PT assesses coagulation factors in extrinsic pathway (F
VII) and common pathway (F X, F V, prothrombin, and
and gradually occlude an aperture at the centre of the
fibrinogen) (Fig. 29.12).
membrane. The time required for complete occlusion of
the aperture is called as closure time. Normal closure Principle: Tissue thromboplastin and calcium are added
time is 1-3 minutes. The PFA-100 test is performed to plasma and clotting time is determined. The test
initially with the collagen/epinephrine membrane; if determines the overall efficiency of extrinsic and
closure time is normal, there is no significant platelet common pathways.
function defect. If closure time with collagen/epine- Equipment
phrine is prolonged, test with collagen/ADP is carried 1. Water bath at 37°C
out; if normal, aspirin-induced platelet dysfunction is the 2. Test tubes (75 × 12 mm)
probable cause; if prolonged, other platelet function 3. Stopwatch
defect (acquired or inherited) is likely. Reagents
This test is more sensitive than bleeding time to assess 1. Thromboplastin reagent: This contains tissue factor
primary hemostasis, sensitive for detection of von and phospholipids and is available commercially.
Willebrand disease and easy to perform. However, in 2. Calcium chloride 0.025 mol/liter.
the presence of thrombocytopenia and anemia, closure Specimen: Platelet-poor citrated plasma (Box 29.2).

tahir99 - UnitedVRG
vip.persianss.ir
Laboratory Tests in Bleeding Disorders 303

Uses of PT
1. To monitor patients who are on oral anticoagulant therapy:
PT is the standard test for monitoring treatment with
oral anticoagulants. Oral anticoagulants inhibit
carboxylation of vitamin K-dependent factors
(Factors II, VII, IX, and X) and make these factors
inactive.
In patients receiving oral anticoagulants, PT
should be reported as a ratio of PT of patient to PT of
control; it should not be reported as percentage.
Various types of thromboplastin reagents obtained
from different sources (like ox brain, rabbit brain, or
rabbit lung) are available for PT test. These differ in
Fig. 29.12: Principle of prothrombin time their responsiveness to deficiency of vit. K-dependent
factors. Technique of PT is also different in different
laboratories. For standardization and to obtain
Box 29.2: Collection of blood for coagulation studies comparable results, it is recommended to report PT
(in persons on oral anticoagulants) in the form of an
Venous blood is collected from antecubital fossa with a
plastic, siliconized glass, or polypropylene syringe and a International Normalized Ratio (INR).
large bore needle (20 or 21 G in adults, 22 or 23 G in infants).
ISI
Blood should never be collected from indwelling intravenous PT of patient
lines, as these often contain heparin. Glass syringe should INR =
PT of control
not be used for collection since it activates coagulation. The
blood is drawn gently but quickly after a single, smooth
venepuncture. The needle is detached from the syringe, and International Sensitivity Index (ISI) of a particular
the sample is passed gently into the plastic container. After tissue thromboplastin is derived (by its manufacturer)
capping the container, the blood and the anticoagulant are by comparing it with a reference thromboplastin of
mixed immediately by gentle inversion 5 times. The known ISI.
anticoagulant used for coagulation studies is trisodium citrate
INR should be maintained in the therapeutic range
(3.2%), with anticoagulant to blood proportion being 1:9.
Most coagulation studies require platelet poor plasma (PPP). for the particular indication (INR of 2.0-3.0 for
To obtain PPP, blood sample is centrifuged at 3000-4000 prophylaxis and treatment of deep venous throm-
revolutions per minute for 15-30 minutes. Coagulation studies bosis; INR of 2.5-3.5 for mechanical heart valves).
are carried out within 2 hours of collection of sample. Therapeutic range provides adequate anticoagulation
for prevention of thrombosis and also checks excess
dosage, which will cause bleeding.
Method
2. To assess liver function: Liver is the site of synthesis of
1. Deliver 0.1 ml of plasma in a glass test tube kept in
water bath at 37°C. various coagulation factors, including vitamin K-
2. Add 0.1 ml of thromboplastin reagent and mix. dependent proteins. Therefore PT is a sensitive test
3. After 1 minute, add 0.1 ml of calcium chloride for assessment of liver function.
solution. Immediately start the stopwatch and record 3. Detection of vitamin K deficiency: PT measures three of
the time required for clot formation. the four vitamin K-dependent factors (i.e. II, VII, and
X).
Normal range: 11-16 seconds.
4. To screen for hereditary deficiency of coagulation factors
Causes of prolongation of PT VII, X, V, prothrombin, and fibrinogen.
1. Treatment with oral anticoagulants
2. Liver disease Activated Partial Thromboplastin Time (APTT)
3. Vitamin K deficiency APTT is a measure of coagulation factors in intrinsic
4. Disseminated intravascular coagulation pathway (F XII, F XI, high molecular weight kininogen,
5. Inherited deficiency of factors in extrinsic and prekallikrein, F IX, and F VIII) and common pathway (F
common pathways. X, F V, prothrombin, and fibrinogen) (Fig. 29.13).

tahir99 - UnitedVRG
vip.persianss.ir
304 Essentials of Clinical Pathology

Fig. 29.13: Principle of activated partial thromboplastin time

Principle: Plasma is incubated with an activator (which Normal range: 30-40 seconds.
initiates intrinsic pathway of coagulation by contact
Causes of prolongation of APTT
activation). Phospholipid (also called as partial thrombo-
1. Hemophilia A or B.
plastin) and calcium are then added and clotting time is 2. Deficiencies of other coagulation factors in intrinsic
measured. and common pathways.
Equipment: This is same as for PT. 3. Presence of coagulation inhibitors
4. Heparin therapy
Reagents 5. Disseminated intravascular coagulation
1. Kaolin 5 gm/liter: This is a contact activator. 6. Liver disease
2. Phospholipid: Various APTT reagents are available
Uses of APTT
commercially, which contain phospholipids.
1. Screening for hereditary disorders of coagulation: Since
3. Calcium chloride 0.025 mol/liter.
deficiencies of F VIII (hemophilia A) and F IX
Specimen: Citrated platelet poor plasma (Box 29.2).
(hemophilia B) are relatively common, APTT is the
Method most important screening test for inherited coagu-
1. Mix equal volumes of phospholipid reagent and lation disorders. APTT detects deficiencies of all
calcium chloride solution in a glass test tube and keep coagulation factors except F VII and F XIII.
in a waterbath at 37°C. PT is also performed along with APTT. Prolongation
2. Deliver 0.1 ml of plasma in another test tube and add of both PT and APTT is indicative of deficiency of
0.1 ml of kaolin solution. Incubate at 37°C in the coagulation factors in common pathway. Normal PT
waterbath for 10 minutes. with prolongation of APTT is indicative of intrinsic
3. After exactly 10 minutes, add 0.2 ml of phospholipid- pathway deficiency (particularly of F VIII or IX).
calcium chloride mixture, start the stopwatch, and 2. To monitor heparin therapy: Heparin potentiates the
note the clotting time. action of natural anticoagulant antithrombin III which

tahir99 - UnitedVRG
vip.persianss.ir
Laboratory Tests in Bleeding Disorders 305

is an inhibitor of thrombin and activated factors IX, X, Thrombin Time (TT)


and XI. Full dose heparin therapy needs monitoring
Thrombin time assesses the final step of coagulation i.e.
by APTT to maintain the dose in the therapeutic range
conversion of fibrinogen to fibrin by thrombin
(1.5 to 2.5 times the upper reference limit of APTT).
(Fig. 29.15).
3. Screening for circulating inhibitors of coagulation: APTT
is prolonged in the presence of specific inhibitors Principle: Thrombin is added to patient’s plasma and time
(which are directed against specific coagulation required for clot formation is noted.
factors) and non-specific inhibitors (which interfere Equipment: Same as for PT.
with certain coagulation reactions).
Reagent: Thrombin solution.
Mixing experiment for detection of inhibitors: Mixing studies
Specimen: Citrated platelet poor plasma (Box 29.2).
are used to distinguish between factor deficiencies and
factor inhibitors (specific coagulation factor inhibitor or Method: Take 0.1 ml of buffered saline in a test tube and
non-specific inhibitor such as lupus anticoagulant). If add 0.1 ml of plasma. Note clotting time after addition
APTT is prolonged, patient’s plasma is mixed with an of 0.1 ml of thrombin solution.
equal volume of normal plasma (called as a 50:50 mix) Normal range: ± 3 seconds of control.
and APTT is repeated. In coagulation factor deficiency,
prolongation of APTT gets corrected by more than 50%
of the difference between the clotting times of control
and test plasma. In the presence of lupus anticoagulant,
there is no such correction. With lupus anticoagulant,
APTT remains prolonged after mixing and for 2 hours
following incubation. With F VIII inhibitor (which is time-
and temperature-dependent), prolonged APTT gets
immediately corrected after mixing, but becomes
prolonged after incubation (Fig. 29.14).
Fig. 29.15: Principle of thrombin time

Fig. 29.14: Interpretation of mixing experiment. Lupus inhibitor is an immediate-acting inhibitor,


while F VIII inhibitor is a time-dependent inhibitor

tahir99 - UnitedVRG
vip.persianss.ir
306 Essentials of Clinical Pathology

Causes of Prolongation of TT 2. Selective prolongation of bleeding time: This occurs


1. Disorders of fibrinogen: Prolongation of TT occurs in in disorders of platelet function, von Willebrand
afibrinogenemia (virtual absence of fibrinogen), disease, and vascular disorders. For definitive
hypofibrinogenemia (fibrinogen less than 100 mgs/dl), diagnosis of platelet function defects, platelet
and dysfibrinogenemia (dysfunctional fibrinogen). aggregation studies are required. In vascular
2. Heparin therapy: Heparin inhibits action of thrombin. disorders, associated clinical and laboratory features
3. Presence of fibrin degradation products (FDPs): These are often suggestive of diagnosis.
interfere with fibrin monomer polymerization. TT is 3. Selective prolongation of APTT: Isolated prolon-
repeated using a mixture of normal plasma and gation of APTT suggests deficiency of coagulation
patient’s plasma. If TT remains prolonged, then FDPs factors in intrinsic pathway, especially that of F VIII
are present (provided patient is not receiving (hemophilia A) or F IX (hemophilia B). Definitive
heparin). diagnosis is based on specific coagulation factor assay.
Deficiency of F XII, high molecular weight kininogen,
Interpretation of Screening Tests or prekallikrein is not associated with clinical
bleeding.
In a patient with a bleeding disorder, results of all the Acquired causes of prolongation of APTT are heparin
screening tests should be interpreted together (Fig. 29.16). therapy and circulating inhibitors of coagulation.
1. Selective thrombocytopenia: If low platelet count is 4. Prolongation of APTT and bleeding time: This
the only abnormality on hemostasis screen, blood combination is observed in von Willebrand disease.
smear and bone marrow examinations are required Platelet aggregation studies are required for definitive
mainly to exclude underlying hematologic disease diagnosis.
like aplastic anemia, leukemia, lymphoma, or 5. Selective prolongation of PT: Isolated prolongation
myelodysplastic syndrome. In the absence of a of PT is suggestive of F VII deficiency. Acquired
hematologic disorder, normal or increased numbers causes, which cause prolongation of only PT, are early
of megakaryocytes in bone marrow is indicative of vitamin K deficiency and start of oral anticoagulant
peripheral destruction of platelets (e.g. idiopathic therapy.
thrombocytopenic purpura, drugs, infections, 6. Prolongation of both PT and APTT: This indicates
collagen disorders). deficiency of one or more factors in common pathway

Fig. 29.16: Evaluation of a suspected bleeding disorder. BT: bleeding time; PC: platelet count;
PT: prothrombin time; APTT: activated partial thromboplastin time

tahir99 - UnitedVRG
vip.persianss.ir
Laboratory Tests in Bleeding Disorders 307

(i.e. F X, F V, prothrombin, or fibrinogen), or laboratories if platelet dysfunction is suspected. These


dysfibrinogenemia. Other causes are heparin therapy, tests are usually indicated in patients presenting with
liver disease, vit K deficiency (in which prolongation mucocutaneous type of bleeding and in whom
of PT is more as compared to APTT), and oral screening tests reveal normal platelet count, prolon-
anticoagulant therapy. ged bleeding time, normal prothrombin time, and
7. All screening tests abnormal: Combination of low normal activated partial thromboplastin time.
platelet count and prolongation of both PT and APTT Platelet aggregation studies are carried out on
is seen in acute disseminated intravascular coagu- platelet-rich plasma using aggregometer. When a
lation (DIC) and liver disease. DIC should be platelet aggregating agent is added to platelet-rich
suspected when a patient with some underlying plasma, platelets form aggregates and optical density
condition (like sepsis, major trauma, malignancy, falls (or light transmission increases); this is recorded
snake bite, obstetric problem, etc.) develops acute by a chart recorder on a strip chart. Commonly used
bleeding manifestations. Other laboratory abnor- platelet aggregating agents are ADP (adenosine 5-
malities in DIC are fragmented red cells on blood diphosphate), epinephrine (adrenaline), collagen,
smear, and raised levels of FDPs, and D-dimer. In arachidonic acid, and ristocetin. ADP (low dose) and
liver disease, in contrast to DIC, fragmented red cells epinephrine induce primary and secondary waves
are absent, D-dimer test is negative, liver function of aggregation (biphasic curve). Primary wave is due
tests are abnormal, and F VIII level is normal. to the direct action of aggregating agent on platelets.
8. All screening tests normal: If, in a patient suspected Secondary wave is due to thromboxane A2 synthesis
of having a bleeding disorder, all the screening tests and secretion from platelets. Collagen, arachidonic
are normal, possibilities include mild forms of von acid and ristocetin induce a single wave of aggre-
gation (monophasic curve) Normal aggregation curve
Willebrand disease, some platelet function defects,
is shown in Figure 29.17. Aggregation patterns in
vascular disorder, mild coagulation factor deficiency,
various platelet function defects are shown in Figures
or F XIII deficiency. Investigations in such cases 29.18 to 29.20, and in Table 29.8.
include blood smear, platelet function studies, 2. Factor VIII assay: One stage F VIII assay based on
workup for von Willebrand disease, and clot APTT is usually performed for diagnosis of F VIII
solubility test for F XIII deficiency. deficiency. Coagulation factor assays are based on the
ability of patient’s plasma to correct specific factor-
Specific Tests for Hemostasis deficient plasma. The abilities of the dilutions of
1. Platelet aggregation studies: Platelet aggregation standard plasma (normal plasma) and patient’s
tests are carried out in specialized hematology plasma to correct the APTT of the plasma known to

Fig. 29.17: Normal platelet aggregation curves

tahir99 - UnitedVRG
vip.persianss.ir
308 Essentials of Clinical Pathology

Fig. 29.18: Platelet aggregation curves in von Willebrand disease and Bernard-Soulier syndrome (absent aggregation
with ristocetin, normal aggregation with ADP, epinephrine, and arachidonic acid)

Fig. 29.19: Platelet aggregation curves in storage pool defect (absent second wave of aggregation with ADP and
epinephrine, absent or greatly diminished aggregation with collagen, and normal ristocetin aggregation)

Fig. 29.20: Platelet aggregation curves in Glanzmann’s thrombasthenia


(absent aggregation with all agonists except ristocetin)

tahir99 - UnitedVRG
vip.persianss.ir
Laboratory Tests in Bleeding Disorders 309

Table 29.8: Laboratory features of platelet function disorders


Disorder Platelet aggregation pattern Other features
Bernard Soulier syndrome Normal with ADP, epinephrine, Autosomal recessive; severe bleeding; giant
collagen, and arachidonic acid; platelets
deficient with ristocetin
Glanzmann’s thrombasthenia Deficient with ADP, epinephrine, Autosomal recessive; severe bleeding;
collagen, and arachidonic acid; small and discrete platelets; defective clot
normal with ristocetin retraction
Storage pool defect Primary wave with ADP and Defects of platelet granules; platelet dense
epinephrine, normal with granules are decreased with deficient
arachidonic acid, deficient with release of ADP, ATP, calcium, and serotonin
collagen, normal with ristocetin
Aspirin-like defect Primary wave with ADP and
epinephrine, deficient with
arachidonic acid, deficient with
collagen, normal with ristocetin
von Willebrand disease Normal with ADP, epinephrine, Autosomal dominant/recessive; abnormality in
collagen, and arachidonic acid; aggregation corrected with cryoprecipitate
deficient with ristocetin

be completely deficient in F VIII are compared. The


results are plotted (clotting times in seconds vs.
percent factor activity) on a log/log graph paper. The
1:10 dilution is considered as 100% activity. F VIII
activity of 50-150% is considered as normal.
3. Detection of fibrinogen/fibrin degradation products
(FDPs): FDPs are fragments produced by proteolytic
digestion of fibrinogen or fibrin by plasmin.
For determination of FDPs, blood is collected in a tube
containing thrombin (to remove all fibrinogen by
converting it into a clot) and soybean trypsin inhibitor
(to inhibit plasmin and thus prevent in vitro
breakdown of fibrin).
A suspension of latex particles linked to anti-
fibrinogen antibodies (or fragments D and E) is mixed
with dilutions of patient’s serum on a glass slide. If
FDPs are present, agglutination of latex particles Fig. 29.21: Principle of latex agglutination test for
occurs (Fig. 29.21). The highest dilution of serum at fibrinogen/fibrin degradation products
which agglutination is detected is used to determine
concentration of FDPs. microparticles coated with monoclonal antibody to D-
Increased levels of FDPs occur in fibrinogenolysis or dimer are mixed with patient’s sample and observed
fibrinolysis. This occurs in disseminated intravascular for microparticle agglutination. As the particle is small,
coagulation, deep venous thrombosis, severe turbidometric endpoint can be determined in auto-
pneumonia, and recent myocardial infarction. mated instruments.
4. Detection of D-dimers: D-dimer is derived from the D-dimer and FDPs are raised in disseminated
breakdown of fibrin by plasmin and D-dimer test is intravascular coagulation, intravascular thrombosis
used to evaluate fibrin degradation. Blood sample can (myocardial infarction, stroke, venous thrombosis,
be either serum or plasma. Latex or polystyrene pulmonary embolism), and during postoperative

tahir99 - UnitedVRG
vip.persianss.ir
310 Essentials of Clinical Pathology

period or following trauma. D-dimer test is commonly • Factors II, V, VII, VIII, IX, X, XI, XII: 50-150%
used for exclusion of thrombosis and thrombotic • vWF:Ag: 50-150%
tendencies.
5. Estimation of fibrinogen: Fibrinogen is commonly CRITICAL VALUES
measured by Clauss method that consists of modifi- • Prothrombin time: > 30 seconds or > 3 times control
cation of thrombin time by diluting plasma; thrombin value
time of diluted plasma is inversely proportional to
• Activated partial thromboplastin time: ≥ 75 seconds
concentration of fibrinogen. Fibrinogen can also be
• Platelet count < 20,000/cmm or > 1 million/cmm
estimated by immunological method; in dysfibrino-
genemia, fibrinogen estimated by functional assay • D-dimer: Positive
(Clauss method) is abnormal while immunological • Plasma fibrinogen: < 100 mg/dl
assay is normal.
6. Platelet glycoprotein analysis: This is done by flow BIBLIOGRAPHY
cytometric analysis for detection of lack of GpIb/IX 1. Evatt BL, Gibbs WN, Lewis SM, McArthur JR.
in Bernanrd Soulier syndrome (deficiency of CD42), Fundamental Diagnostic Hematology: The Bleeding
and lack of GpIIb/IIIa in Glanzmann’s thromb- and Clotting Disorders (2nd ed), 1992. US Dept. of
asthenia (deficiency of CD41, CD61). health and Human Services, Atlanta, Georgia and
World Health Organization, Geneva, Switzerland.
REFERENCE RANGES 2. Kawthalkar SM. Essentials of Hematology. New Delhi:
Jaypee Brothers Medical Publishers (P) Ltd, 2006.
• Bleeding time: Ivy method: 2-7 minutes; Template 3. Lewis SM, Bain BJ, bates I (Eds). Dacie and Lewis
method: 2.5-9.5 minutes Practical Hematology (9th ed). London: Churchill
• Prothrombin time: 11-16 seconds Livingstone, 2002.
4. Provan D, Krentz A. Oxford Handbook of Clinical and
• Activate partial thromboplastin time: 30-40 seconds Laboratory Investigations (2002). Oxford university
• Thrombint time: ±3 seconds of control Press. Oxford.
• Plasma fibrinogen: 200-400 mg/dl 5. Shrikhande AV, Warhadpande MS, Kawthalkar SM.
• Fibrinogen/fibrin degradation products: < 10 μg/ml A laboratory manual of coagulation (1994). Dept. of
Pathology. Govt. Medical College, Nagpur.
• D-dimer: Qualitiative: Negative; Quantitative: < 200 6. Wallach J. Interpretation of Diagnostic Tests (7th ed).
mg/L Philadelphia: Lippincott Williams and Wilkins, 2000.

tahir99 - UnitedVRG
vip.persianss.ir
30
Laboratory Tests in
Thrombophilia

Thrombophilia is a hemostatic disorder in which there removal of F Va leads to increased prothrombinase


is a predisposition to thrombosis due to an inherited or complex (F Xa-Va-PL-Ca) activity, and greater
acquired condition (Table 30.1). It results from alteration thrombin generation. It is primarily associated with
in hemostatic constituents of blood. increased risk of venous thrombosis. Heterozygotes
have 2-6% increased risk, while homozygotes have
INHERITED THROMBOPHILIA 50-100 times risk. Risk of thrombosis is greatly
1. Factor V Leiden (Activated protein C resistance): increased during pregnancy and following oral
Factor V Leiden (so-called because of its discovery in contraceptive use.
Leiden, Netherlands) is the most common inherited Laboratory tests for FV Leiden include activated
cause of thrombophilia. Mutation in FV gene protein C resistance assay and genetic analysis. In
(transmitted through autosomal dominant inheri- activated protein C resistance assay, activated partial
tance) leads to replacement of arginine by glutamine thromboplastin time (APTT) on the patient’s sample
at position 506. This substitution renders activated is compared with APTT done after addition of
form of FV resistant to degradation by activated activated protein C to plasma sample. The ratio is
protein C. More than 95% cases of activated protein called as activated protein C sensitivity ratio. In
C resistance are due to FV Leiden (other causes of normal subjects, addition of activated protein C to
activated protein C resistance being pregnancy, oral the plasma sample will prolong APTT because
contraceptive use, malignancy, and other FV activated protein C inhibits F V and F VIII. In F V
mutations). The terms FV Leiden and activated Leiden, APTT shows only slight prolongation. Low
protein C resistance are not synonymous. Failure of activated protein C ratio indicates more resistance to

Table 30.1: Conditions associated with increased risk of thrombosis


Inherited (Primary) Acquired (Secondary)
1. Factor V Leiden (Activated protein C resistance), i.e. 1. Oral contraceptive therapy
Abnormal F V that resists degradation by protein C
2. Prothrombin gene mutation 2. Pregnancy
3. Hyperhomocysteinemia 3. Antiphospholipid syndrome
4. Deficiency or decreased activity of protein C 4. Paroxysmal nocturnal hemoglobinuria
5. Deficiency or decreased activity of protein S 5. Malignancy
6. Deficiency or decreased activity of antithrombin III 6. Disseminated intravascular coagulation
7. Dysfibrinogenemia 7. Heparin-induced thrombocytopenia
8. Thrombotic thrombocytopenic purpura
9. Nephrotic syndrome
10. Myeloproliferative disorders

tahir99 - UnitedVRG
vip.persianss.ir
312 Essentials of Clinical Pathology

activated protein C. If the screening test is positive for increased synthesis of coagulation factors in liver and
activated protein C resistance, genetic testing is decreased synthesis of antithrombin III.
performed. Genetic analysis by polymerase chain 2. Antiphospholipid syndrome: Antiphospholipid
reaction can detect heterozygous and homozygous antibodies are autoantibodies directed against
states. antigens composed of phospholipids. They are of two
2. Prothrombin gene G20210A mutation: A G→A types: lupus anticoagulant (so named because it was
mutation at position 20210 of prothrombin gene is first detected in a patient with systemic lupus
the second most common cause of inherited thrombo- erythematosus) and anticardiolipin antibodies. Lupus
philia. This mutation, by some unknown mechanism, anticoagulant is detected by prolongation of phos-
leads to a rise in the concentration of prothrombin, pholipid-dependent coagulation tests such as
thus making available large amounts of prothrombin activated partial thromboplastin time (APTT) and
for conversion to thrombin. It is implicated in dilute Russell’s viper venom time. If APTT is
prolonged, the test is repeated after mixing the sample
causation of both arterial and venous thrombosis and
50:50 with normal plasma. If not corrected, it suggest
pregnancy-associated thrombosis. Diagnosis is based
presence of lupus anticoagulant (Box 30.1). Anti-
on genetic analysis.
cardiolipin antibodies are detected by enzyme linked
3. Deficiency of protein C and S: Protein C, when
immunosorbent assay (ELISA). Antiphospholipid
activated by thrombin, inactivates factors V and VIII.
antibodies are associated with arterial and venous
Protein S serves as a cofactor in this reaction. In thrombosis, spontaneous, recurrent abortions, and
protein C and S deficiency, thrombin generation is thrombocytopenia. Antiphospholipid syndrome is
increased, producing hypercoagulability. Both present if antiphospholipid antibodies (lupus
protein C and protein S deficiencies primarily cause anticoagulant or anticardiolipin antibodies or both)
venous thrombosis. Diagnosis of protein C deficiency are present along with an episode of arterial or venous
requires quantification of protein C concentration. thrombosis, thrombocytopenia, or frequent second
Distinction between deficiency and dysfunction of trimester abortions. Antiphospholipid antibodies
protein C is based on functional assay for protein C. may occur without any underlying disorder
Protein S deficiency is detected by quantification of (primary) or in association with systemic lupus
protein S (both free and bound forms). erythematosus, Sjogren’s syndrome, rheumatoid
4. Deficiency of antithrombin III: Antithrombin III is arthritis, human immunodeficiency virus infection or
the natural inhibitor of thrombin, F Xa, IXa, XIa, and malignancy (secondary).
XIIa. Deficiency state is associated with venous 3. Heparin-induced thrombocytopenia: This compli-
thrombosis. Homozygous state is incompatible with cation occurs in 1-3% of patients receiving any type
life. of heparin. Platelet count should be checked every
5. Hyperhomocysteinemia: Increased levels of the alternate day in patients receiving heparin. The
amino acid homocysteine can occur in various condition should be suspected when patient is not
inherited disorders like homocystinuria, cystathione
synthase deficiency, and C677T gene polymorphism
Box 30.1: Criteria for diagnosis of lupus anticoagulant
in the methyl tetrahydrofolate reductase (MTHFR) gene. (International Society of Thrombosis and Hemostasis)
Acquired causes of hyperhomocysteinemia are vitamin
B12 deficiency, folate deficiency, and vitamin B6 • Prolongation of a phospholipid-dependent screening test
deficiency. Hyperhomocysteinemia is associated with for coagulation like activated partial thromboplastin time
(APTT) or dilute Russell’s viper venom time
increased risk of both arterial and venous thrombosis.
• Mixing study with APTT using 50:50 mixture of patient’s
Laboratory tests are assay for homocyseine or genetic
and normal plasma shows no correction, indicating that
analysis of MTHFR gene. prolongation is due to an inhibitor.
• Demonstration of antiphospholipid nature of antibodies
ACQUIRED THROMBOPHILIA by neutralizing them with high concentration of platelets
(platelet neutralization test)
1. Oral contraceptive therapy and pregnancy: Increa- • No other cause for thrombosis
sed predisposition to thrombosis results from

tahir99 - UnitedVRG
vip.persianss.ir
Laboratory Tests in Thrombophilia 313

responding to heparin, and develops thrombo- 5. Recurrent abortions (≥ 3)


cytopenia (50% below baseline level) and thrombosis. 6. Pregnancy-associated thrombosis.
These patients are at risk of developing acute
myocardial infarction, stroke, peripheral arterial Laboratory Tests
thrombosis and deep venous thrombosis. The The tests should be performed atleast 4-6 weeks after
pathogenesis consists of binding of IgG-heparin- the acute thrombotic event and discontinuation of
platelet factor 4 complexes to Fc receptors on platelets warfarin, as acute states cause elevation of acute phase
that causes activation and aggregation of platelets as reactants (that interfere with testing and cause false
well as clearance of coated platelets by macrophages. positive results).
This leads to thrombosis and thrombocytopenia. 1. For inherited thrombophilia:
• DNA analysis for F V Leiden
LABORATORY TESTING IN THROMBOPHILIA
• DNA analysis for prothrombin gene mutation
Laboratory studies will identify the underlying cause in • Test for homocysteine level.
majority of patient presenting with thrombosis. The • Testing for protein C, protein S, and antithrombin
screening tests for thrombophilia are not readily III deficiency or dysfunction
available. Also, testing for thrombophilia is expensive, 2. For acquired thrombophilia:
rarely helps in acute patient management, and results • Exclusion of diabetes mellitus, hyperlipidemia,
may not be interpreted correctly, leading to improper myeloproliferative disorders, paroxysmal noctur-
treatment. Concentration solely on thrombophilia nal hemoglobinuria
detection can overlook a serious underlying disorder. • Test for lupus anticoagulant and anticardiolipin
Testing for thrombophilia should be ordered in selected antibodies.
patients in whom such complex and costly testing will
be of significant benefit for management. BIBLIOGRAPHY
1. Brandt JT, Triplett DA, Alving B, et al. Criteria for the
Indications for Testing for Thrombophilia diagnosis of lupus anticoagulants: an update. Thromb
Hemost 1995;74:1185-90.
1. Thrombosis at young age with no obvious risk factors. 2. Van Cott EM, Laposata M. Laboratory evaluation of
2. Unexplained recurrent thrombosis. hypercoagulable states. Hematology/Oncology Clinics
3. Thrombosis at unusual locations. of North America 1998;12:1141-66.
3. Whiteman T, Hassouna HI. Hypercoagulable states.
4. Strong family history of thrombosis (in first degree Hematology/Oncology Clinics of North America 2000;
relative). 14:355-77.
31
Laboratory Tests in
Porphyrias

Porphyrias (from Greek porphura meaning purple various steps as shown in Figure 31.1. Each of the steps is
pigment; the name is probably derived from purple catalyzed by a separate enzyme; if any of these steps fails
discoloration of some body fluids during the attack) are (due to hereditary or acquired cause), precursors of heme
a heterogeneous group of rare disorders resulting from (porphyrin intermediates) accumulate in blood, get
disturbance in the heme biosynthetic pathway leading deposited in skin and other organs, and excreted in urine
to the abnormal accumulations of red and purple and feces. Depending on the site of defect, different types
pigments called as porphyrins in the body. Heme, a of porphyrias are described with varying clinical features,
component of hemoglobin, is synthesized through severity, and the nature of accumulated porphyrin.

Fig. 31.1: Figure on left shows steps in the biosynthesis of heme. Some steps (first and last three) occur in mitochondria,
while some occur in cytosol. Figure on right shows deficiency state associated with each enzyme. Deficiency of ALA synthase
is associated with sideroblastic anemia, and deficiencies of other enzymes cause porphyria
Laboratory Tests in Porphyrias 315

Porphyria has been offered as a possible explanation precursors up to the point of enzyme defect. Increased
for the medieval tales of vampires and werewolves; this levels of heme precursors cause symptoms of acute
is because of the number of similarities between the porphyria. When the heme level returns back to normal,
behavior of persons suffering from porphyria and the symptoms subside.
folklore (avoiding sunlight, mutilation of skin on Accumulation of porphyrin precursors can occur in
exposure to sunlight, red teeth, psychiatric disturbance, lead poisoning due to inhibition of enzyme aminolevulinic
and drinking of blood to obtain heme). acid dehydratase in heme biosynthetic pathway. This can
Porphyrias are often missed or wrongly diagnosed mimick acute intermittent porphyria.
as many of them are not associated with definite physical
findings, screening tests may yield false-negative results,
CLINICAL FEATURES
diagnostic criteria are poorly defined and mild disorders
produce an enzyme assay result within ‘normal’ range. Clinical features of porphyrias are variable and depend
Heme is mainly required in bone marrow (for hemo- on type. Acute porphyrias present with symptoms like
globin synthesis) and in liver (for cytochromes). acute and severe abdominal pain/vomiting/consti-
Therefore, porphyrias are divided into erythropoietic and pation, chest pain, emotional and mental disorders,
hepatic types, depending on the site of expression of seizures, hypertension, tachycardia, sensory loss, and
disease. Hepatic porphyrias mainly affect the nervous muscle weakness. Cutaneous porphyrias present with
system, while erythropoietic porphyrias primarily affect photosensitivity (redness and blistering of skin on
the skin. Porphyrias are also classified into acute and non- exposure to sunlight), itching, necrosis of skin and gums,
acute (or cutaneous) types depending on clinical and increased hair growth over the temples (Table 31.2).
presentation (Table 31.1). Symptoms can be triggered by drugs (barbiturates,
Inheritance of porphyrias may be autosomal domi- oral contraceptives, diazepam, phenytoin, carbama-
nant or recessive. Most acute porphyrias are inherited in zepine, methyldopa, sulfonamides, chloramphenicol,
an autosomal dominant manner (i.e. inheritance of one and antihistamines), emotional or physical stress,
abnormal copy of gene). Therefore, the activity of the infection, dieting, fasting, substance abuse, premenstrual
deficient enzyme is 50%. When the level of heme falls in period, smoking, and alcohol.
the liver due to some cause, activity of ALA synthase is Autosomal dominant porphyrias include acute
stimulated leading to increase in the levels of heme intermittent porphyria, variegate porphyria, porphyria

Table 31.1: Various classification schemes for porphyrias


Classification based on Classification based on site of Classification based on
predominant clinical expression of disease mode of clinical
manifestations presentation

Neuropsychiatric Hepatic Acute


1. Acute intermittent porphyria 1. ALA-dehydratase porphyria 1. ALA-dehydratase porphyria
(Plumboporphyria)
2. ALA-dehydratase porphyria 2. Acute intermittent porphyria 2. Acute intermittent
(Plumboporphyria) porphyria
Cutaneous (Photosensitivity) 3. Hereditary coproporphyria 3. Hereditary coproporphyria
1. Congenital erythropoietic 4. Variegate porphyria 4. Variegate porphyria
porphyria
2. Porphyria cutanea tarda Erythropoietic porphyria Non-acute (cutaneous)
3. Erythropoietic protoporphyria 1. Congenital erythropoietic 1. Porphyria cutanea tarda
porphyria
Mixed (Neuropsychiatric and cutaneous) 2. Erythropoietic protoporphyria 2. Congenital erythropoietic
porphyria
1. Hereditary coproporphyria Hepatic/Erythropoietic 3. Erythropoietic protoporphyria
2. Variegate porphyria 1. Porphyria cutanea tarda
316 Essentials of Clinical Pathology

Table 31.2: Clinical characteristics of porphyrias


Porphyria Deficient enzyme Clinical features Inheritance Initial test
1. Acute intermittent PBG deaminase Acute neurovisceral Autosomal dominant Urinary PBG; urine
porphyria attacks; triggering becomes brown,
(AIP)* factors+ (e.g. drugs, red, or black on
diet restriction) standing
2. Variegate Protoporphyrinogen Acute Autosomal Urinary PBG
porphyria oxidase neurovisceral dominant
attacks + skin
fragility, bullae
3. Hereditary Coproporphyrinogen Acute Autosomal Urinary PBG
coproporphyria oxidase neurovisceral dominant
attacks + skin
fragility, bullae
4. Congenital Uroporphyrinogen Onset in infancy; Autosomal Urinary/fecal
erythropoietic cosynthase skin fragility, recessive total porphyrins;
porphyria bullae; extreme ultraviolet
photosensitivity fluorescence of
with mutilation; urine, feces,
red teeth and urine and bones
(pink red urine-
staining of diapers)
5. Porphyria Uroporphyrinogen Skin fragility, Autosomal Urinary/fecal
cutanea tarda* decarboxylase bullae dominant total porphyrins
(some cases)
6. Erythropoietic Ferrochelatase Acute Autosomal Free erythrocyte
protoporphyria* photosensitivity dominant protoporphyrin

Disorders marked with * are the three most common porphyrias. PBG: Porphobilinogen

cutanea tarda, erythropoietic protoporphyria (most cases), be submitted for detection of excessive urinary excretion
and hereditary coproporphyria. Autosomal recessive of porphobilinogen (PBG) (Fig. 31.2). In AIP, urine becomes
porphyrias include: congenital erythropoietic porphyria, red or brown on standing (Fig. 31.3). In suspected cases of
erythropoietic protoporphyria (few cases), and ALA- cutaneous porphyrias (acute photosensitivity without skin
dehydratase porphyria (plumboporphyria). fragility), free erythrocyte protporphyrin or FEP in EDTA
blood (for diagnosis of erythrocytic protoporphyria) and
LABORATORY DIAGNOSIS for all other cutaneous porphyrias (skin fragility and
Porphyria can be diagnosed through tests done on blood, bullae), examination of fresh, random urine (10-20 ml)
urine, and feces during symptomatic period. Timely and and either feces (5-10 g) or plasma for excess porphyrins
accurate diagnosis is required for effective management are necessary (Fig. 31.4 and Table 31.2).
of porphyrias. Due to the variability and a broad range of Apart from diagnosis, the detection of excretion of a
clinical features, porphyrias are included under particular heme intermediate in urine or feces can help
differential diagnosis of many conditions. All routine in detecting site of defect in porphyria. Heme precursors
hospital laboratories usually have facilities for initial up to coproporphyrinogen III are water-soluble and thus
investigations in suspected cases of porphyrias; laboratory can be detected in urine. Protoporphyrinogen and
tests for identification of specific type of porphyrias are Protoporphyrin are insoluble in water and are excreted
available in specialized laboratories. in bile and can be detected in feces.
All samples should be protected from light. Samples
Initial Studies
required are (i) 10-20 ml of fresh random urine sample
In suspected acute porphyrias (acute neurovisceral attack), without any preservative; (ii) 5-10 g wet weight of fecal
a fresh randomly collected urine sample (10-20 ml) should sample, and (iii) blood anticoagulated with EDTA.
Laboratory Tests in Porphyrias 317

Fig. 31.2: Evaluation of acute neurovisceral porphyria

Fig. 31.3: Red coloration of urine on


standing in acute intermittent porphyria

Fig. 31.4: Evaluation of cutaneous porphyrias


318 Essentials of Clinical Pathology

Table 31.3: Diagnostic patterns of concentrations of heme precursors in acute porphyrias


Porphyria Urine Feces

Acute intermittent porphyria PBG, Copro III –


Variegate porphyria PBG, Copro III Proto IX
Hereditary coproporphyria PBG, Copro III Copro III
PBG: Porphobilinogen; Copro III: Coproporphyrinogen III; Proto IX: Protoporphyrin IX

Table 31.4: Diagnostic patterns of concentrations of heme precursors in cutaneous porphyrias


Porphyria Urine Feces Erythrocytes

Congenital erythropoietic porphyria Uro I, Copro I Copro I –


Porphyria cutanea tarda Uroporphyrin Isocopro –
Erythropoietic protoporphyria – – Protoporphyrin
Uro I: Uroporphyrinogen I; Copro I: Coproporphyrinogen I; Isocopro: Isocoproporphyrinogen

Test for Porphobilinogen in Urine Tests for Porphyrins in Erythrocytes and Plasma
Ehrlich’s aldehyde test is done for detection of PBG. Visual examination for porphyrin fluorescence, and
Ehrlich’s reagent (p-dimethylaminobenzaldehyde) reacts solvent fractionation and spectrophotometry have now
with PBG in urine to produce a red color. The red product been replaced by fluorometric methods.
has an absorption spectrum with a peak at 553 nm and a Further Testing
shoulder at 540 nm. Since both urobilinogen and
If the initial testing for porphyria is positive, then
porphobilinogen produce similar reaction, further testing
concentrations of porphyrins should be estimated in
is required to distinguish between the two. Urobilinogen
urine, feces, and blood to arrive at specific diagnosis
can be removed by solvent extraction. (See Watson- (Tables 31.3 and 31.4)
Schwartz test in Chapter 1: Examination of Urine). In latent porphyrias and in patients during remission,
Levels of PBG may be normal or near normal in porphyrin levels may be normal; in such cases, enzymatic
between attacks. Therefore, samples should be tested and DNA testing is necessary for diagnosis.
during an attack to avoid false-negative results. If porphyria is diagnosed, then it is necessary to
investigate close family members for the disorder.
Test for Total Porphyrins in Urine
Positive family members should be counseled regarding
Total porphyrins can be detected in acidified urine triggering factors.
sample by spectrophotometry (Porphyrins have an
intense absorbance peak around 400 nm). Semi- BIBLIOGRAPHY
quantitative estimation of porphyrins is possible. 1. Deacon AC, Elder GH: Frontline tests for the investi-
Test for Total Porphyrins in Feces gation of suspected porphyria. J Clin Pathol 2001; 54;
500-7.
Total porphyrins in feces can be determined in acidic 2. Crook MA: Clinical chemistry and metabolic medicine.
extract of fecal sample by spectrophotometry; it is Seventh edition. London. Edward Arnold, 2006.
necessary to first remove dietary chlorophyll (that also 3. Thadani H, Deacon A, Peters T: Diagnosis and
absorbs light around 400 nm) by diethyl ether extraction. management of porphyria. BMJ 2000; 320; 1647-51.
32

Automation in Hematology

AUTOMATED HEMATOLOGY ANALYZER Automated hematology analyzers are of two main types:
• Semi-automated: Some steps like dilution of blood
Automation is a process of replacement of tasks hitherto
sample are performed by the technologist; can
performed by humans by computerized methods.
measure only a few parameters
Until recently, hematological tests were performed
• Fully automated: Require only anticoagulated blood
only by manual methods. These methods, though still
sample; measure multiple parameters.
performed in many peripheral laboratories, are labor-
intensive, and involve use of hemocytometers (counting
chambers), centrifuges, Wintrobe tubes, photometers, PRINCIPLES OF WORKING
and stained blood smears. Hematology cell analyzers can Automated hematology analyzers work on different
generate the blood test results rapidly and also perform principles:
additional tests not possible by manual technology. • Electrical impedance
Both manual and automated laboratory techniques • Light scatter
have advantages and disadvantages, and it is unlikely • Fluorescence
that one will completely replace the other. • Light absorption
• Electrical conductivity.
Advantages of Automated Hematology Analyzer
Most analyzers are based on a combination of
• Speed with efficient handling of a large number of different principles.
samples 1. Electrical impedance: This is the classic and time-
• Accuracy and precision in quantitative blood tests tested technology for counting cellular elements of
• Ability to perform multiple tests on a single platform blood. As this method of cell counting was first
• Significant reduction of labor requirements developed by Coulter Electronics, it is also called as
• Invaluable for accurate determination of red cell Coulter principle (Fig. 32.1). Two electrodes placed
indices. in isotonic solutions are separated by a glass tube
having a small aperture. A vacuum is applied and as
Disadvantages of Automated a cell passes through the aperture, flow of current is
Hematology Analyzer
impeded and a voltage pulse is generated.
• Flags: Flagging of a laboratory test result demands The requisite condition for cell counting by this
labour-intensive manual examination of a blood method is high dilution of sample so that minimal
smear numbers of cells pass through the aperture at one
• Comments on red cell morphology cannot be point of time. There are two electrodes on either side
generated. Abnormal red cell shapes (such as of the aperture; as the solution in which the cells are
fragmented cells) cannot be recognized. suspended is an electrolyte solution, an electric
• Erroneously increased or decreased results due to current is generated between the two electrodes.
interfering factors When a cell passes through this narrow aperture
• Expensive with high running costs. across which a current is flowing, change in electrical
320 Essentials of Clinical Pathology

while those between 36-360 fl are counted as red cells.


Hemoglobin is estimated by light transmission at 535
nm.
2. Light scatter: Each cell flows in a single line through
a flow cell. A laser device is focused on the flow cell;
as the laser light beam strikes a cell it is scattered in
various directions. One detector captures the forward
scatter light (forward angle light scatter or FALS) that
is proportional to cell size and a second detector
captures side scatter (SS) light (90°) that corresponds
to the nuclear complexity and granularity of
cytoplasm. This simultaneous measurement of light
scattered in two directions is used for distinguishing
Fig. 32.1: Coulter principle of electrical impedance between granulocytes, lymphocytes, and monocytes.
3. Fluorescence: Cellular fluorescence is used to
measure RNA (reticulocytes), DNA (nucleated red
resistance (i.e. momentary interruption of electrical
cells), and cell surface antigens.
current between the two electrodes) occurs. A small
4. Light absorption: Concentration of hemoglobin is
pulse is generated due to a temporary increase in
measured by absorption spectrophotometry, after
impedance. This pulse is amplified, measured, and conversion of hemoglobin to cyanmethemoglobin or
counted. The height of the pulse is proportional to some other compound. In some analyzers, peroxidase
cell volume. The width of the pulse corresponds with cytochemistry is used to classify leukocytes; the
the time required for the cell to traverse the aperture. peroxidase activity is determined by absorbance.
Cells that do not pass through the center of the 5. Electrical conductivity: Some analyzers use
aperture generate a distorted pulse that is not conductivity of high frequency current to determine
representative of the cell volume. Some analyzers use physical and chemical composition of leucocytes for
hydrodynamic focusing to force the cells through the their classification.
central path so that all cells take the same path for
volume measurement. PARAMETERS MEASURED BY
An anticoagulated whole blood sample is aspirated HEMATOLOGY ANALYZERS
into the system, divided into two portions, and mixed
with a diluent. One dilution is passed to the red cell Parameters measured by hematology analyzers and their
aperture bath (for red cell and platelet counting), and derivation are shown in Tables 32.1 and 32.2. Most
the other is delivered to the WBC aperture bath automated hematology analyzers measure red cell count,
(where a reagent is added for lysis of red cells and red cell indices (mean cell volume, mean cell hemoglobin,
release hemoglobin; this portion is used for leukocyte mean cell hemoglobin concentration), hemoglobin,
counting followed by estimation of hemoglobin). hematocrit, total leukocyte count, differential leukocyte
Particles between 2-20 fl are counted as platelets, count (three-part or five-part), and platelet count.

Table 32.1: Parameters measured by hematology analyzers


Parameters measured by most analyzers Parameters measured by some analyzers

• RBC count • Red cell distribution width


• Hemoglobin • Reticulocyte count
• Mean cell volume • Reticulocyte hemoglobin content
• Mean cell hemoglobin • Mean platelet volume
• Mean cell hemoglobin concentration • Platelet distribution width
• WBC count • Reticulated platelets
• WBC differential
• Platelet count
Automation in Hematology 321

Table 32.2: Parameters reported by hematology analyzers

Parameters measured directly or Parameters measured through


derived through histogram calculation

• RBC count • Hematocrit


• Mean cell volume (Derived from RBC histogram) • Mean cell hemoglobin
• Red cell distribution width (Derived from RBC histogram) • Mean cell hemoglobin concentration
• Hemoglobin
• Reticulocyte count
• WBC count
• Differential WBC count (Derived through WBC histogram)
• Platelet count
• Mean platelet volume (Derived from platelet histogram)

Estimation of Hemoglobin
Hemoglobin is measured directly by a modification of
cyanmethemoglobin method (all hemoglobins are
converted to cyanmethemoglobin by potassium
ferricyanide; cyanmethemoglobin has a broad absor-
bance peak at 540 nm). Some analyzers use a non-
hazardous reagent such as sodium lauryl sulphate. A
non-ionic detergent is added for rapid red cell lysis and
to minimize turbidity caused by cell membranes and
plasma lipids.
Fig. 32.2: Diagrammatic representation of red cell histogram
Estimation of Red Blood Cell Count and obtained by aperture impedance. The analyzer counts cells
Mean Cell Volume (MCV) between 36 fl and 360 fl as red cells. Although leukocytes
are present and counted along with red cells in the diluting
Red cell count and cell volume are directly measured by fluid, their number is not statistically significant. Only if
aperture impedance or light scatter analysis. In a red cell leukocyte count is markedly elevated (>50,000/μl), histogram
histogram, cell numbers are plotted on Y-axis, while cell and the red cell count will be affected. Area of the peak
volume is indicated on X-axis (Fig. 32.2). The analyzer between 60 fl and 125 fl is used for calculation of mean
counts those cells as red cells volume of which ranges cell volume and red cell distribution width. Abnormalities in
between 36 fl and 360 fl. red cell histogram include: (1) Left shift of the curve in
MCV is used for morphological classification of microcytosis, (2) Right shift of the curve in macrocytosis, and
anemia into microcytic, macrocytic, and normocytic (3) Bimodal peak of the curve in double (dimorphic)
types. population of red cells

Estimation of MCH, MCHC, and Hematocrit


Mean cell volume (fl)
These parameters are obtained indirectly through Hematocrit (%) = ———————————
calculations. Red cell count (106/μl)
Hemoglobin (g/l)
Mean cell hemoglobin (pg) = ——————————— Estimation of Red Cell Distribution Width (RDW)
Red cell count (106/μl) RDW is a quantitative measure of variation in sizes of
red cells and is expressed as coefficient of variation of
Hemoglobin (g/dl) red cell size distribution. It is equivalent to anisocytosis
Mean cell hemoglobin = ——————————— observed on blood smear. It is derived from red cell
concentration (g/dl) Hematocrit (%) histogram in some analyzers. RDW is usually elevated
322 Essentials of Clinical Pathology

in iron deficiency anemia, but not in β-thalassemia minor Instruments measuring a 5-part differential work on a
and anemia of chronic disease (other causes of microcytic combination of different principles, e.g. light scatter,
anemia). However, this distinction is not absolute and impedance, and electrical conductivity, a combination
there is a significant overlap between values among of light scatter, peroxidase staining, and resistance of
patients. Raised RDW requires examination of blood basophils to lysis in acid buffer, etc.
smear.
Among the red cell values generated by the analyzer Platelet Count
(red cell count, hemoglobin, hematocrit, MCV, MCH, Platelets are difficult to count because of their small size,
MCHC, and RDW), most important for decision- marked variation in size, tendency to aggregation, and
making are hemoglobin, hematocrit, and MCV. overlapping of size with microcytic red cells, cellular
fragments, and other debris. In hematology analyzers,
WBC Differential
this difficulty is addressed by mathematical analysis of
Hematology analyzers can either generate a 3-part platelet volume distribution so that it corresponds to log-
differential (differential count reported as lymphocytes, normal distribution. Platelets are counted by electrical
monocytes, and granulocytes) or a 5-part differential impedance method in the RBC aperture, and a histogram
(lymphocytes, monocytes, neutrophils, eosinophils, and is generated with platelet volume on X-axis and relative
basophils). The 3-part differential counting is based on cell frequency on Y-axis (Fig. 32.4). Normal platelet
electrical impedance volume measurement of leukocytes. histogram consists of a right-skewed single peak.
In volume histogram for WBCs, approximate numbers Particles greater than 2 fl and less than 20 fl are classified
of cells are plotted on Y-axis and cell size on X-axis. Those as platelets by the analyzer.
cells with volume 35-90 fl are designated as lymphocytes, Two other platelet parameters can be obtained from
cells with volume 90-160 fl as mononuclear cells, and platelet histogram using computer technology: mean
cells with volume 160-450 fl as neutrophils (Fig. 32.3). platelet volume (MPV) and platelet distribution width
Any deviation from the expected histogram is flagged (PDW). Some analyzers can generate another parameter
by the analyzer, mandating review of blood smear. A called as reticulated platelets.
large proportion of 3-part differential counts are ‘flagged’ MPV refers to the average size of platelets and is
to avoid missing abnormal cells. obtained from mathematical calculation. Normal MPV

Fig. 32.3: Diagrammatic representation of WBC histogram. WBC histogram analysis shows relative numbers of cells on
Y-axis and cell size on X-axis. The lytic agent lyses the cytoplasm that collapses around the nucleus causing differential
shrinkage. The analyzer sorts the WBCs according to the nuclear size into 3 main groups (3-part differential): Cells with
35-90 fl volume are designated as lymphocytes, cells with 90-160 fl volume are designated as monocytes, and cells with
160-450 fl volume are designated as neutrophils. Abnormalities in WBC histogram include: (1) Peak to the left of lymphocyte
peak: Nucleated red cells, (2) Peak between lymphocytes and monocytes: Blast cells, eosinophilia, basophilia, plasma
cells, and atypical lymphocytes, and (3) Peak between monocytes and neutrophils: Left shift
Automation in Hematology 323

graph are determined by the degree of side scatter, degree


of forward scatter, light absorption by the cell, and
cytochemical staining (if used). The forward angle light
scatter (FALS) is represented on Y-axis, and the side
scatter (SS) is represented on X-axis. Low FALS and low
SS are indicative of lymphocytes; with subsequent
increasing FALS and SS, monocytes, neutrophils, and
lastly eosinophils are designated in the graph. Counting
of basophils is based on a different technology.
Fig. 32.4: Diagrammatic representation of normal platelet
histogram: Counting and sizing of platelets by electrical FLAGGING
impedance method occurs in the RBC aperture. The counter
designates particles between sizes 2 fl and 20 fl as platelets. ‘Flags’ are signals that occur when an abnormal result is
Abnormalities in platelet histogram result from interferences detected by the analyzer. Flags are displayed to reduce
such as cytoplasmic fragments (peak at left end of histogram) false-positive and false-negative results by mandating a
or severely microcytic red cells and giant platelets (peak at
review of blood smear examination.
right end of histogram)

CAUSES OF ERRONEOUS RESULTS


(INTERFERENCES CAUSING ABNORMAL
is 7-10 fl. Increased MPV (> 10 fl) results from presence of RESULT)
immature platelets in circulation; peripheral destruction
These are listed in Table 32.3.
of platelets stimulates megakaryocytes to produce such
platelets (e.g. in idiopathic thrombocytopenic purpura).
Decreased MPV (< 7 fl) is due to presence of small platelets
FLOW CYTOMETRY
in circulation (in conditions associated with reduced Flow cytometry is a procedure used for measuring
production of platelets in bone marrow). multiple cellular and fluorescent properties of cells when
PDW is analogous to RDW and is a measure of variation they flow as a single cell suspension through a laser beam
in size of platelets (normal <20%). Increased PDW is by a specialized instrument called as a flow cytometer.
observed in megaloblastic anemia, chronic myeloid Flow cytometry can analyze numerous cells in a short
leukemia, and after chemotherapy. time and multiple parameters of a single cell can be
Some analyzers measure reticulated platelets or analyzed simultaneously. From the measured para-
young platelets that contain RNA (similar to reticulo- meters, specific cell populations are defined. Cells or
cytes). Increased numbers of reticulated platelets are seen particles with size 0.2-150 μm are suitable for flow
in thrombocytopenia due to peripheral destruction of cytometer analysis.
platelets. Flow cytometry can provide following information
about a cell (Box 32.1):
Reticulocyte Count
• Cell size (forward scatter)
Various fluorescent dyes can combine with RNA of • Internal complexity or granularity (side scatter)
reticulocytes; the fluorescence then is counted in a flow • Relative fluorescence intensity.
cytometer. More immature reticulocytes fluoresce more A flow cytometer consists of three main components
strongly as they contain more RNA. or systems: fluidics, optics, and electronics.
Reticulocyte hemoglobin content is a parameter that 1. Fluidics: The function of the fluidics system is to
estimates hemoglobinization of most recently produced transport cells in a stream to the laser beam for
red cells. It is a predictor of iron deficiency. interrogation. Cells (fluorescence-tagged) are
introduced into the cytometer (injected into the sheath
WBC Cytogram (Scattergram) fluid within the flow chamber) and made to flow in a
In the scattergram, each dot represents a cell of a given single file past a laser (light amplification by
volume and density, and the positions of dots in the stimulated emission of radiation) beam. The stream
324 Essentials of Clinical Pathology

Table 32.3: Causes of erroneous results with hematology analyzer


Parameter Interfering factors

Erroneous increase Erroneous decrease


1. All parameters — • Clotted sample
1. WBC count • Nucleated red cells • Clotted sample
• Large platelet clumps
• Unlysed red cells (some abnormal
red cells resist lysing)
• Cryoglobulins
2. RBC count • Very high WBC* • Clotted sample
• Large numbers of giant platelets • Microcytic red cells
• Autoagglutination
3. Hemoglobin • Very high WBC • Clotted sample
• Hyperlipidemia
• High bilirubin
4. MCV • Very high WBC • Cryoglobulins
• Hyperglycemia
• Autoagglutination (cold agglutinins)
5. MCHC • Hyperlipidemia • Very high WBC
• Autoagglutination (cold agglutinins)
6. Platelets • Microcytic red cells • Platelet satellitism
• WBC fragments • Platelet clumping
• Cryoglobulins
*: WBCs are counted along with RBCs, but normally their number is statistically insignificant

are then collected by appropriately positioned lenses.


Box 32.1: Properties of a cell measured by a A system of optical mirrors and filters then directs
flow cytometer
the specified wavelengths of light to the designated
• Relative size detectors. Two types of light signals are generated
• Relative granularity or internal complexity when the laser beam strikes cells tagged with
• Relative fluorescence
fluorescent molecules: fluorescence and light scatter.
The cells tagged with fluorescence emit a momentary
pulse of fluorescence; in addition, two types of light
transporting the cells should be positioned in the
scatter are measured: forward scatter (proportional
center of the laser beam. The portion of the fluid
to cell diameter) and side scatter (proportional to
stream where the cells are located is called as the
granularity of cell).
sample core. Only a single cell or particle should pass
3. Electronics: The optical signals (photons) are
through the laser beam at one time. Flow cytometers
converted to corresponding electronic signals
use the principle of hydrodynamic focusing (process
(electrons) by the photodetectors (photodiodes and
of centering the sample core within the sheath fluid)
for presenting cells to the laser. photomultiplier tubes). The electronic signal pro-
2. Optics: This system consists of lasers for illumination duced is proportional to the amount of light striking
of cells in the sample, and filters to direct the a cell. The electric current travels to the amplifier and
generated light signals to the appropriate detectors. is converted to a voltage pulse. The voltage pulse is
The light source used in most flow cytometers is assigned a digital value representing a channel by
laser. The laser most commonly used in flow the Analog-to Digital Converter (ADC). The channel
cytometry is Argon-ion laser. The light signals are number is then transferred to the computer which
generated when the laser beam strikes the cell, which displays it to the appropriate position on the data plot.
Automation in Hematology 325

TERMINOLOGY IN FLOW CYTOMETRY When a cell passes through laser beam, side scatter
and fluorescent signals that are emitted by the cell are
Fluorescence
directed to photomultiplier tubes, while the forward scatter
A fluorochrome absorbs light energy and emits excess signals are directed to a photodiode. Photomultiplier tubes
energy in the form of photon light (fluorescence). and photodiodes are called as detectors. Optical filters
Fluorescence is the property of molecules to absorb light are placed before the detectors that allow only a narrow
at one wavelength and emit light at a longer wavelength. range of wavelengths to reach the detectors (Fig. 32.5).
The fluorescent dyes commonly used in flow cytometry
are fluorescein isothiocyanate (FITC) and phycoerythrin Data Analysis
(PE). The fluorochrome-labeled antibodies are used for
detection of antigenic markers on the surface of cells. A The data collected and stored in the computer can be
particular cell type can be identified on the basis of the displayed in various formats. A parameter means forward
antigenic profile expressed. Multiple fluorochromes can scatter, or side scatter, or emitted fluorescence from a
be used to identify different cell types in a mixed particle as it passes through a laser beam. A histogram is
population of cells. a data plot of a single parameter, with the parameter’s
signal value in channel numbers or relative fluorescence
Light Scatter intensity on X-axis (horizontal axis) and number of events
Light is scattered when the incident light is deflected by a on the Y-axis. A dot plot is a two parameter data graph in
particle traversing through a beam of light. This depends which each dot represents one event that the flow
on the physical properties of the cell. Two forms of light cytometer analyzed; one parameter is displayed on the X-
scatter are used to identify different cell types: forward axis and the other on the Y-axis. A 3-D plot represents one
scatter and side scatter. Forward scatter (light scattered in parameter on X-axis, another parameter on Y-axis, and
the same direction as the laser beam) is related to cell size. number of events per channel on Z-axis.
Side scatter (light scattered at a 90° angle to the laser beam)
Gating
is related to internal granularity of the cell. Main
subpopulations of leukocytes are identified on the A gate is a boundary that can be set to restrict the analysis
basis of correlated measurements of forward and side to a specific population within the sample. For example, a
scatters. gate boundary can be drawn on a dot plot or histogram to

Fig. 32.5: Principle of working of a flow cytometer


326 Essentials of Clinical Pathology

restrict the analysis only to cells with the size of 2. Paroxysmal nocturnal hemoglobinuria: Deficiency of
lymphocytes. Gates can be inclusive (selection of events CD 55 and CD 59.
that fall within the boundary) or exclusive (selection of 3. Hematopoietic stem cell transplantation: Enumeration
events that fall outside the boundary). Data selected by
of CD34+ stem cells.
the gate is then displayed in subsequent plots.
4. Feto-maternal hemorrhage: Detection and quantitation
Sorting of foetal hemoglobin in maternal blood sample.
5. Anemias: Reticulocyte count.
Usually, when a cell passes through the laser beam, it is
6. Human immunodeficiency virus infection: For
sent to waste. Sorting consists of collecting cells of interest
(defined through criteria of size and fluorescence) for enumeration of CD4+ lymphocytes.
further analysis (such as microscopy or functional or 7. Histocompatibility cross matching.
chemical analysis). Sorting feature is available only in
some flow cytometers. BIBLIOGRAPHY
COMMON APPLICATIONS OF FLOW 1. Henry JB. Clinical Diagnosis and Management by
CYTOMETRY IN HEMATOLOGY Laboratory Methods (20th Ed). Philadelphia: WB
Saunders Company, 2001.
1. Leukemias and lympomas: Immunophenotyping 2. Houwen B. The differential count. Laboratory Hema-
(evaluation of cell surface markers), diagnosis, tology 2001;7:89-100.
detection of minimal residual disease, and to identify 3. Ward PJ. The CBC at the turn of the millennium: an
prognostically important subgroups. overview. Clin Chem 2000;46:1215-20.
Practical Blood Transfusion
33

Blood Group Systems

International Society of Blood Transfusion (ISBT) Table 33.1: Blood group systems
Working Party has organized red cell antigens into 25 ABO Dombrock
blood group systems (Table 33.1). Red cell antigens that MNS Colton
are produced by alleles (alternative forms of a specified P Landseiner-Weiner
gene) at a single gene locus or very closely linked loci Rh Chido/Rogers
constitute a blood group system. Lutheran Hh
Kell Kx
Blood group genes are inherited in a Mendelian
Lewis Gerbich
manner and are mostly located on autosomes. Most of Duffy Cromer
the blood group genes are expressed in a co dominant Kidd Knops
manner (i.e. the two allelic forms are expressed equally Diego Indian
if inherited in a heterozygous state, and no gene or Yt Ok
allele is dominant over another). The particular alleles Xg Raph
Scianna
at a specified gene locus in an individual constitute the
genotype. Phenotype is the outward expression of the
genotype. ABO SYSTEM
In blood transfusion practice, most important blood In ABO system, there are four main types of blood groups:
group systems are ABO and Rh. This is because, A, B, A, B, AB, and O. Identification of these four blood groups
and Rh D antigens are the most immunogenic (i.e. is based on presence or absence of A and/or B antigens
capable of eliciting a strong antibody response on on red cells. According to Landsteiner’s law, anti-A and/
stimulation) and their alloantibodies can cause destruc- or anti-B antibodies are always present in plasma of
tion of transfused red cells or induce hemolytic disease individuals who lack corresponding antigen(s) on their
of newborn (HDN). ABO antigens are also important in red cells (Table 33.2). ABO is the only blood group system,
organ transplantation. in which if an antigen is absent in an individual,

Table 33.2: ABO blood groups


Blood group Caucasian Indian Genotype Antigen(s) Antibody in plasma
frequency frequency on red cells

A 40% 27% AA or AO A Anti-B


B 11% 31% BB or BO B Anti-A
AB 4% 8% AB AB Nil
O 45% 34% OO Nil Anti-A and anti-B

Note: A and B genes are dominant while O gene is recessive. Individuals with AA, BB, and OO are homozygous
respectively for A, B, and O, while AO, BO, and AB are heterozygous.
330 Essentials of Clinical Pathology

corresponding antibody is always present in plasma.


Box 33.1: Distribution of ABH antigens
There is a geographical variation in frequencies of
different ABO blood groups. However, generally, O • Red blood cells
blood group is the most common, while AB group is the • White blood cells (weak)
least common in a population. • Platelets (weak)
• Body tissues
• Body fluids (in secretors)
Blood Group O
This is usually the most frequently occurring blood group
in the population. Group O red cells have large amounts secretors) (Box 33.1).
of H antigen on their surface. Genotype is OO and the ABO antigens are poorly expressed at birth, increase
antibodies in plasma are anti-A, anti-B, and anti-A,B. gradually in strength and become fully expressed around
These antibodies are usually IgG in nature. They can 1 year of age. In older age, they become slightly weak.
therefore cross the placenta and induce hemolytic disease
Formation of ABH Antigens
of newborn.
The H gene (genotype HH or Hh) produces a transferase
Blood Group A enzyme, which changes precursor substance (present on
Genotype of group A persons is either AA or AO, and red cells) into H substance. The A and B genes (chromo-
the antigens present on the red cells are A and H. Their some 9) produce specific transferase enzymes, which
plasma contains anti-B antibodies that are IgM in nature. convert H substance into A and B antigens respectively.
Blood group A is divided into two subgroups: A1 (80%) Sugar N-acetylgalactosamine is added to H antigen chain
and A2 (20%). Anti-A1 produced by A2 or A2B to make the A antigen, while sugar galactose is added to
individuals is weak and clinically insignificant; however, H antigen chain to produce the B antigen. Some amount
it can cause problems during blood grouping. Some of H antigen remains unchanged on red cells. The O gene
persons with A2 and with A2B can make anti-A1. A1 produces an inactive transferase so that H antigen
and A2 red cells can be differentiated by lectins of Dolichos persists unchanged on red cells (Fig. 33.1).
biflorus that cause agglutination of A1 red cells, but not Some persons do not inherit the H gene (genotype
of A2 red cells. hh) and thus cannot synthesize H substance. Such persons
may inherit the A or B gene but cannot express it, as they
Blood Group B are unable to produce the H substance. Such individuals
are said to have Bombay phenotype or Bombay blood
Genotype of group B persons is either BB or BO, and the group (Oh) (the genotype of Bombay phenotype is hh).
antigens present on red cells are B and H. Their plasma The name derives its name because it was first detected
contains anti-A antibodies that are IgM in nature. in persons living in Bombay, India. It has also been
described in other populations. This blood group is
Blood Group AB
extremely rare. There is complete lack of H, A, and B
This is the least common of the ABO blood groups. antigens due to the lack of H (and Se) genes. Their red
Genotype is AB, and antigens present on red cells are A, cells type as group O; however, unlike group O
B, and very small amount of H. The AB blood group is individuals, Oh persons have no H antigen on their red
further subdivided into A1B and A2B. Plasma of group cells and their plasma contains strong anti-H in addition
AB individuals contain no anti-A or anti-B antibodies. to anti-A and anti-B. On cross-matching, all units are
incompatible (Box 33.2). Therefore Bombay group
Antigens of the ABO system persons should be transfused only with Oh blood.
Antigens of the ABO system are: A (A1, A2), B, and H.
Secretors and Non-secretors
They are glycolipids, glycoproteins, or glycosphingo-
lipids. In addition to red cells, they are also expressed on Secretors are persons who secrete A, B, and H antigens
white cells, platelets, and body tissues. They are also into body fluids (such as plasma, gastric juice, saliva,
present in a soluble form in various body secretions (in sweat, tears, semen, milk, etc.). This ability is dependent
Blood Group Systems 331

Fig. 33.1: Formation of antigens of the ABO system

Box 33.2: Identification of Bombay phenotype not match with the ABO blood group of the accused).
Inhibitor tests are used to detect the presence of soluble
• Blood group: O blood group antigens in body secretions. If saliva contains
• Cross-matching: Serum incompatible with O cells a soluble antigen, and if corresponding antibody is
• Red cells yield negative reaction with anti-H lectin
added, the activity of the antibody is neutralized. When
red cells carrying the appropriate antigens are subse-
on presence of a dominant secretor gene (Se) on quently added to the mixture, there will be inhibition of
chromosome 19. About 80% of individuals are secretors agglutination (i.e. the person is a secretor). If aggluti-
(genotype Sese or SeSe) and remaining are non-secretors nation occurs, then the individual is a non-secretor.
(genotype sese). Both secretors and non-secretors express Example:
ABO antigens on red cells. 1. Blood group: B
Antigens secreted by different ABO blood groups are: Saliva + anti-B; Add B cells: No agglutination
• Group A: A, H Interpretation: Secretor
• Group B: B, H 2. Blood group: O
• Group AB: A, B, H Saliva + Anti-A; Add A cells: Agglutination; Saliva +
• Group O: H Anti-B; Add B cells: Agglutination; Saliva + Anti-H;
Antigens secreted into body fluids are called as ABH Add O cells: No agglutination.
substances. Testing for ABH substances in saliva may be Interpretation: Secretor
helpful when red cell grouping yields uncertain results. 3. In non-secretors, no soluble antigens are present in
Determining secretor status in saliva and semen can be secretions. Thus antibodies in the reagent are free to
helpful in resolving ABO blood group discrepancies and bind to red cells of respective group. There will be
in forensic studies (e.g. semen sample collected from a agglutination reaction.
rape victim revealing a soluble ABH antigen that does
332 Essentials of Clinical Pathology

Antibodies of the ABO System B antibodies. Therefore, group O persons are traditionally
considered as universal donors.Group AB persons are
The most important antibodies in transfusion practice are
considered as universal recipients. This, however, is an
anti-A and anti-B. They are also called as naturally
occurring antibodies because they arise without immune oversimplification because only the reaction between
recipient’s plasma (antibodies) and the donor’s red cells
stimulation by red cells by relevant blood group antigens.
is considered; a small amount of antibodies in donor’s
They are regularly-occurring, i.e. if an antigen is absent
plasma can cause destruction of recipient’s red cells. In
the corresponding antibody is always present. They are
addition, antigens other than A, B, or RhD can bind to
not detectable in the blood of newborn infants due to
corresponding antibodies or immunize the recipient.
their underdeveloped immune system and appear
around 3-6 months of life. It is thought that they are
THE Rh SYSTEM
produced in response to A- and B-like antigens of
bacteria, which are present in the intestine and certain When Rhesus monkey red cells were injected into rabbits
foods. If anti-A and/or anti-B are present at birth, they and guinea pigs (by Landsteiner and Weiner in 1940),
are of maternal origin (IgG). Anti-A and anti-B antibodies antibody, which was raised, was found to react with
are usually of IgM class. Immune or IgG antibodies (anti- Rhesus monkey red cells as well as with 85% of human
A or anti-B) can be stimulated by exposure to red cells, red cells (White residents of New York city). The antigen
white cells, or platelets. involved was called as Rh factor. Subsequently it was
IgM antibodies are large molecules and can bind up shown that the original antibody was different from anti-
to ten antigens; therefore they can cause direct D antibody discovered later. The name of the antigen,
agglutination of red cells and cause visible agglutination. however, has remained as Rhesus. Apart from
IgM antibodies can efficiently fix the complement. Landsteiner and Weiner, credit for discovery of Rh
Naturally occurring ABO antibodies can cause: system also goes to Levine and Stetson who discovered
• Hemolytic transfusion reaction in a case of ABO- in 1939 the antibody (actually anti-D) that caused
mismatched blood transfusion, hemolytic disease of newborn.
• Acute graft rejection in case of ABO-incompatible The Rh system is only next in importance to ABO
solid organ transplantation, and system in transfusion practice. The importance of this
• Hemolysis of donor red cells following ABO- system lies in the high immunogenicity of Rh D antigen,
incompatible bone marrow transplantation. which readily induces formation of anti-D antibodies in
Less commonly, some individuals have large 50-70% of Rh D-negative individuals. Anti-D antibodies
amounts of ABO antibodies of immune nature. Usually, can cause hemolytic transfusion reaction or, in pregnant
group O individuals following immune stimulation by women, Rh hemolytic disease of newborn.
transfusion, pregnancy, or injection of certain vaccines
or toxoids (that contain bacterial A- and B-like antigens) The Rh system was discovered independently by:
produce them in large amounts. These antibodies are of (1) Stetson and Levine in 1939 and (2) by Landsteiner
IgG class, of high titer, and cannot be neutralized by and Weiner in 1940.
soluble blood group antigens. If blood of such group O
individuals (called dangerous universal group O donors)
is transfused to group A or B individuals, serious Antigens of the Rh System
hemolysis of recipient’s red cells can occur. Therefore Rh system is highly complex and consists of about 40
group O donors should not be employed as universal antigens. The important antigens of the Rh system are
donors. In addition to causing hemolytic transfusion C, D, E, c, and e. Antigen d does not exist. D antigen is
reaction, these IgG antibodies can cross the placenta and the most immunogenic. There are various nomenclature
induce hemolytic disease of newborn. systems for Rh antigens. Fisher-Race or CDE nomen-
clature system and Weiner system are popular.
Concepts of Universal Donor and Recipient
According to Fisher and Race, three closely linked
Red cells of group O donors are devoid of A and B genes are inherited together on one chromosome
antigens and cannot be agglutinated by anti-A and anti- (haplotype) from each parent. Allelic forms of these genes
Blood Group Systems 333

are C and c, D and d, and E and e with eight possible In Rh negative persons, deletions, point mutations, or
haplotypes: Cde, cde, cDE, cDe, cdE, Cde, CDE, and CdE. partial mutations of D gene have been found. Rh antigens
As an individual inherits one haplotype from each are expressed only on red cells and not on any other
parent, 36 genotypes are possible such as Cde/cde, Cde/ tissues. They are also not secreted in body fluids. In
cDe, CDE/cde, etc (Fig. 33.2). The presence of D in either contrast to ABO antigens, Rh antigens are fully expressed
homozygous (D/D) or heterozygous (D/d) state makes on red cells before birth and also on red cells of early
that individual Rh positive, while Rh negative persons fetuses.
are homozygous for d (d/d). It was thought that d gene Depending on the presence or absence of antigen D
was an amorph. on red cells, a person is grouped either as Rh positive
According to the theory by Dr. Alexander Weiner, a (when red cells express antigen D) or Rh negative (when
single Rh gene is inherited from each parent; this single D antigen is absent on red cells). Frequency of D antigen
Rh gene, however, has multiple alleles (Fig. 33.3). The varies in different populations. In India, approx. 95% of
Weiner system uses Rh-Hr nomenclature. The major the people express D antigen on their red cells (Rh D
difference between Fisher-Race and Weiner systems is positive), while 5% are Rh D negative. The frequency
that according to Fisher-Race, there are three closely in Caucasians is 85% Rh positive and 15% Rh negative.
linked genes that are inherited from each parent, while Other forms of D antigen are weak D and partial D
according to Weiner, a single gene with multiple alleles (Fig. 33.5). Red cells having weak D antigen were
is inherited from each parent. formerly called as Du cells which react weakly with anti-
Results of current genetic studies suggest that both D reagent. There is a quantitative reduction in the
Fisher-Race and Weiner systems are partially correct. It number of D antigen sites on such red cells. Du recipients
has been found that the RH locus is located on do not make anti-D antibodies following stimulation by
chromosome 1 and consists of two closely linked genes- D antigen (e.g. following D positive blood transfusion).
RHD and RHCE (Fig. 33.4). The alleles of RHCE are CE, Du donors should be considered as Rh positive and their
Ce, ce, and cE. blood should not be transfused to Rh negative donors.

Fig. 33.2: Fisher-Race system of nomenclature of Rh blood group inheritance


334 Essentials of Clinical Pathology

Fig. 33.3: Weiner theory of Rh system inheritance

Fig. 33.4: According to current genetic studies, there are two genes on
short arm of chromosome 1: RHCE and RHD

Fig. 33.5: Diagrammatic representation of Fig. 33.6: Relative immunogenicity of Rh antigens. D antigen
variations of Rh antigen is the most immunogenic while e antigen is the weakest
Blood Group Systems 335

Table 33.3: Blood group systems other than ABO and Rh


Blood group Antigens Antibodies Comment
system

1. Lewis Lea, Leb Natural, IgM Lewis antigens are passively absorbed from plasma on red cells;
Lewis antibodies are rarely of clinical significance
2. Kell About 20 IgG Anti-K antibodies can cause hemolytic transfusion reaction and
(KEL1, hemolytic disease of newborn; Some individuals do not have
KEL2, etc) precursor substance on their red cells from which K antigens are
produced; such red cells have short life, acanthocytic features, and
express Kell antigens weakly (MacLeod phenotype).
3. Duffy Fya, Fyb IgG Antibodies can cause hemolytic transfusion reaction. Plasmodium
vivax enters the red cells at the Duffy antigen site. The Fy(a-b-)
phenotype in blacks confers resistance against Plasmodium vivax
infection
4. Kidd JKa, JKb IgG Antibodies cause delayed transfusion reaction and mild hemolytic
disease of newborn
5. MNSs M, N, S, s M and N antigens are important in paternity testing
k
6. P P, P1, P IgM, IgG Auto-anti-P occurs in paroxysmal cold hemoglobinuria

In red cells having partial D antigen, parts of D antigen be transfused only with Rh-negative blood. During
are missing. Variants of partial D antigen exist. pregnancy, IgG anti-D can cross the placenta and induce
Individuals with DVI variant are able to produce anti-D hemolytic disease of newborn by causing immune
antibody against the missing part of the antigen. Such hemolysis of fetal red cells. Rh hemolytic disease of
recipients should be considered as Rh negative, while newborn can be prevented by prophylactic adminis-
donors should be regarded as Rh positive. However, in
tration of Rh immune globulin to all Rh-negative women
practice, individuals with partial D antigen are typed as
during mid pregnancy and within 72 hours of delivery.
D negative and are identified only after they have
Anti-D and anti-c can cause severe HDN. Anti-C, anti-
produced anti-D antibodies.
Complete absence of all Rh antigens on red cells E, and anti-e usually do not cause HDN or cause mild
(Rh null cells) is associated with stomatocytosis and HDN. Relative immunogenicity of Rh antigens is shown
compensated hemolysis. in Figure 33.6.

OTHER BLOOD GROUP SYSTEMS


Rh Antibodies
Salient features of some other blood group systems are
In general, most Rh antibodies are of immune type, i.e. summarized in Table 33.3.
they are the result of immunization by blood transfusion
BIBLIOGRAPHY
or pregnancy. Most of these antibodies are of IgG class.
Rh antibodies can cause hemolytic transfusion 1. Cheesbrough M. District Laboratory Practice in Tropical
Countries. Part 1 and Part 2. Cambridge: Cambridge
reaction or hemolytic disease of newborn (HDN). Since University Press, 1998.
Rh antibodies do not activate complement, hemolysis is 2. Kawthalkar SM. Essentials of Hematology. New Delhi:
Jaypee Brothers Medical Publishers (P) Ltd, 2006.
extravascular and predominantly occurs in spleen. Due
3. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical
to the high immunogenicity of D antigen, Rh negative Hematology (9th ed). London: Churchill Livingstone,
persons (especially women of childbearing age) should 2001.
34

Blood Grouping

ABO GROUPING
Box 34.1: ABO antisera
There are two methods for ABO grouping:
• Three types: Anti-A, anti-B, anti-A, B.
• Cell grouping (forward grouping): Red cells are tested
• Anti-A: Blue-colored;
for the presence of A and B antigens employing
• Anti-B: Yellow-colored.
known specific anti-A and anti-B (and sometimes
• Anti-A, B: Colorless.
anti-A, B) sera.
• Serum grouping (reverse grouping): Serum is tested for • Sodium azide is added to prevent the growth of bacteria
the presence of anti-A and anti-B antibodies by • Antisera are kept stored at 4-6°C to preserve their potency
employing known group A and group B reagent red • Routinely, anti-A and anti-B are used for blood grouping.
cells. • Indications for using anti-A, B: (1) for confirmation of cell
grouping in newborns, and (2) to resolve ABO group
Both cell and serum grouping should be done since
discrepancies.
each test acts as a check on the other.
• Antisera may be polyclonal or monoclonal. Monoclonal
There are three methods for blood grouping: tube, antisera are specific, avid, and can detect weak antigens.
microplate, and slide. Slide test is described below. Tube Monoclonal antisera are commonly used.
and microplate methods are better and are employed in
blood banks; they are outlined in short following slide
test.

SLIDE TEST
Principle
Red cells from the specimen are reacted with reagent
antisera (anti-A and anti-B). Agglutination of red cells
indicates presence of corresponding antigen (aggluti-
nogen) on red cells.

Specimen
Capillary blood from finger prick, or venous blood
collected in EDTA anticoagulant.

Reagents
ABO antisera: See box 34.1 and Figure 34.1. Fig. 34.1: Anti-A and anti-B sera used for cell grouping

Method 2. Place one drop of anti-A serum and one drop of anti-
1. A clean and dry glass slide is divided into two sections B serum in the center of the corresponding section of
with a glass marking pencil. The sections are labeled the slide. Antiserum must be taken first to ensure that
as anti-A and anti-B to identify the antisera (Fig. 34.2). no reagents are missed.
Blood Grouping 337

Table 34.1: Interpretation of cell grouping


(forward grouping) by slide test
Anti-A Anti-B Blood group
+ – A
– + B
+ + AB
– – O

Slide test is quick and needs only simple equipment. It


can be used in blood donation camps and in case of an
emergency. However, it is not recommended as a routine
test in blood banks since weakly reactive antigens on cells
on forward grouping and low titer anti-A and anti-B on
reverse grouping may be missed. Also, drying of the
reaction mixture at the edges causes aggregation that may
be mistaken for agglutination. Results of slide test should
always be confirmed by cell and serum grouping by tube
method.

Fig. 34.2: Cell grouping by slide method TUBE METHOD


Test tube method is more reliable than slide test, but takes
3. Add one drop of blood sample to be tested to each longer time and more equipment. For cell grouping,
drop of antiserum. patient’s saline-washed red cells are mixed with known
4. Mix antiserum and blood by using a separate stick or antiserum in a test tube; the mixture is incubated at room
a separate corner of a slide for each section over an temperature, and centrifuged. For serum grouping,
patient’s serum is mixed with reagent red cells of known
area about 1 inch in diameter.
group (available commercially or prepared in the
5. By tilting the slide backwards and forwards, examine
laboratory), incubated at room temperature, and
for agglutination after exactly two minutes.
centrifuged (Table 34.2). Following centrifugation, a red
6. Result: cell button (sediment) will be seen at the bottom of the
Positive (+): Little clumps of red cells are seen floating tube. Cell button is dislodged by gently tapping the base
in a clear liquid. of the tube and examined for agglutination.

Negative (–): Red cells are floating homogeneously in a Positive (+) Test
uniform suspension. Clumps of red cells suspended in a clear fluid. Aggluti-
7. Interpretation: Interpret the result as shown in the nation in tube test is graded from 1+ to 4+ and read
Table 34.1 and Figure 34.2. macroscopically (Fig. 34.3).

Table 34.2: Interpretation of forward (cell) and reverse (serum) grouping


Forward (Cell) grouping Reverse (serum) grouping Interpretation
Anti-A serum Anti-B serum A1 cells B cells Blood group
+ – – + A
– + + – B
– – + + O
+ + – – AB

+: Agglutination of red cells; –: No agglutination of red cells.


338 Essentials of Clinical Pathology

Test tube method of blood grouping is more reliable


than slide method. This is because centrifugation
enhances the reaction by bringing antigen and antibodies
closer together and allows detection of weaker antigen-
antibody reactions; in addition drying is avoided and
smaller amounts of reagent are required.

MICROPLATE METHOD
Microplate is a polystyrene plate consisting of 96 micro
wells of either U- or V-shape. Grouping is carried out in
micro wells. This method is sensitive and ideal for large
number of samples (Fig. 34.4).

FALSE REACTIONS IN ABO GROUPING


Fig. 34.3: Grading of ABO tube test. Negative: Uniform 1. Autoagglutination: Presence of IgM autoantibodies
suspension of red cells; Grade 1 (1+): Many small clumps reactive at room temperature in patient’s serum can
of red cells (fine granular appearance); Grade 2 (2+): Many lead to autoagglutination. If autocontrol is not used,
large clumps with many free red cells; Grade 3 (3+): Three
blood group in such a case will be wrongly typed as
or four individual clumps with few free red cells; and Grade
4(4+): One solid clump of red cells with no free red cells AB. Therefore, for correct result, if autocontrol is also
showing agglutination, cell grouping should be
repeated after washing red cells with warm saline,
Negative (–) Test
and serum grouping should be repeated at 37°C.
Uniform suspension of red cells. 2. Rouleaux formation: Rouleux formation refers to red
Separate tubes of auto-control, positive control, and cells adhering to each other like a stack of coins and
negative control should always be setup along with the can be mistaken for agglutination. Rouleaux forma-
test sample tube.
tion is caused by high levels of fibrinogen, immuno-
Auto-control tube consists of mixture of patient’s red globulins, or intravenous administration of a plasma
cells and patient’s own serum. This is required to rule
expander such as dextran. Rouleaux formation (but
out false-positive result due to autoantibodies in patient’s
not agglutination) can be dispersed by addition of
serum causing auto-agglutination of patient’s own red
normal saline during serum grouping.
cells. Auto-control test is particularly essential when ABO
3. False-negative result due to inactivated antisera: For
grouping is being done only by forward method and
preservation of potency of antisera, they should be
blood group is typed as AB. If there are autoantibodies
kept stored at 4°-6°C. If kept at room temperature for
in recipient’s serum, ABO grouping, Rh typing, antibody
long, antisera are inactivated and will give false-
screening, and cross-matching all will show positive
result. negative result.
4. Age: Infants start producing ABO antibodies by 3-6
In two positive control tubes, anti-A serum is mixed
months of age and serum grouping done before this
with group A red cells and anti-B is mixed with group B
red cells respectively. In two negative control tubes, anti- age will yield false-negative result. Elderly indivi-
A serum is mixed with group B red cells and anti-B serum duals also have low antibody levels.
is mixed with group A red cells respectively. These
controls are necessary to confirm that reagents are Rh D GROUPING
working properly. D antigen is the most immunogenic after ABO antigens
and therefore red cells are routinely tested for D.
If forward grouping, reverse grouping, and autocontrol
Individuals are called as Rh-positive or Rh-negative
tests are all positive, then these results are probably
depending on presence or absence of D antigen on their
indicative of a cold-reactive autoantibody. Before
red cells. Following transfusion of Rh-positive blood to
performing forward typing, red cells should be washed
Rh-negative persons, 70% of them will develop anti Rh-
with normal saline to elute the antibody. Before
performing reverse grouping, autoantibody should be D antibodies. This is of particular importance in women
of childbearing age as anti-D antibodies can crosss the
adsorbed by washed cells till autocontrol is negative.
placenta during pregnancy and destroy D-positive fetal
Blood Grouping 339

Fig. 34.4: Microplate (96 well) method for blood grouping. Numbers 1 to 12 represent patient identification numbers. Reagents
added to the patient sample are written on left, while interpretation (blood group of patient) is written at the bottom. For understanding,
reaction patterns of the 8 possible blood groups are shown, while the last 4 columns have been kept empty. Red compact
button indicates agglutination, while uniform suspension indicates no agglutination

Table 34.3: Comparison of ABO grouping and Rh typing


Parameter ABO grouping Rh typing

1. Blood groups A, B, AB, O Positive, Negative


2. Forward grouping Yes Yes
3. Reverse grouping Yes No
4. Antisera IgM IgG or IgM
5. Dosage effect* No Yes
6. Color coding of antisera A:Blue, B: Yellow, AB: Colorless D: Colorless
7. Optimum reaction temperature Room temperature 37°C or room temperature

*In dosage effect, antibody reacts stronger with homozygous cells than with heterozygous cells

red cells and cause hemolytic disease of newborn. In other Rh typing is done at the same time as ABO grouping.
sensitized individuals, re-exposure to D antigen can Method of Rh D grouping is similar in principle to ABO
cause hemolytic transfusion reaction. grouping. Since serum or reverse grouping is not possible,
In Rh D grouping, patient’s red cells are mixed with each sample is tested in duplicate. Dosage effect (stronger
anti-D reagent. Serum or reverse grouping is not carried antigen-antibody reaction in homozygous cells i.e.
out because most Rh-negative persons do not have anti- stronger reaction with DD) is observed with antigens of
D antibodies; anti-D develops in Rh-negative individuals the Rh system. Autocontrol (patient’s red cell + patient’s
only following exposure to Rh-positive red cells. serum) and positive and negative controls are included
340 Essentials of Clinical Pathology

in every test run. Monoclonal IgM anti-D antiserum BIBLIOGRAPHY


should be used for cell grouping, which allows Rh 1. Cheesbrough M. District Laboratory Practice in Tropical
grouping to be carried out at the same time as ABO Countries. Part 1 and Part 2. Cambridge: Cambridge
grouping at room temperature. With monoclonal University Press, 1998.
antisera, most weak and variant forms of D antigen are 2. Henry JB. Clinical Diagnosis and Management by
detected and further testing for weak forms of D antigen Laboratory Methods. (20th Ed). Philadelphia: WB
(Du) is not required. Saunders Company, 2001.
Differences between ABO and Rh grouping are 3. Kawthalkar SM. Essentials of Hematology. New Delhi:
shown in Table 34.3. Jaypee Brothers Medical Publishers (P) Ltd, 2006.
35

Collection of Donor Blood,


Processing and Storage

In India, collection of blood, processing, storage, and blood donation. General steps for collection of donor blood
preparation of blood components and derivatives are are shown in Figure 35.1. The safe transfusion practice
regulated by Food and Drugs Administration (FDA). starts with proper selection of blood donors; if donors are
Blood is regarded as a “drug” (under section 3(b) of the not properly screened, blood can become a medium of
Drugs and Cosmetics Act, 1940) by the FDA and all the transmission of infections like human immunodeficiency
blood banks have to obtain a license from the FDA and virus, hepatitis B and C virus, syphilis, and malaria.
follow the FDA guidelines. Blood donations are of three main types: whole blood
donation (collection of one unit or 350 ml of whole blood
TYPES OF BLOOD DONORS in an anticoagulant solution), autologous donation
(donation for subsequent transfusion to self), and
Blood donation is a process through which a blood donor apheresis donation (removal of whole blood from a
has a specified amount of blood drawn for the purpose donor, separation and retention of the desired portion,
of storage in a blood bank and for subsequent blood and returning the remaining portion to the donor).
transfusion. Blood donation may be done in a blood bank There are three main types of whole blood donors:
or in blood donation camps. The process of blood • Voluntary
donation involves selection of blood donors by screening, • Professional
actual donation of blood, and a brief recovery period after • Replacement.

Fig. 35.1: General steps before collection of donor blood


342 Essentials of Clinical Pathology

A blood bank should function only on voluntary blood Box 35.1: Typical questions in the
donations. A voluntary blood donor donates blood out of questionnaire asked to the prospective donor
his/her own free will and on humanitarian grounds or
out of sense of duty or responsibility towards the • Have you donated blood previously? If yes, date of last
community. A voluntary donor is issued a voluntary donor donation and discomfort during or after donation if any.
• Were you deferred as a donor?
card which can be presented for free blood unit if blood is
• Did you receive any blood or blood products in the last
required for himself or a close family member. 6 months?
Paid or professional donors donate blood for money. • Do you currently suffer from or were suffering from any
As payment encourages concealment of a significant of the following conditions: Heart disease, lung disease,
illness, high-risk behavior, or medical history, such form liver disease, kidney disease, cancer, diabetes, epilepsy,
of donations should be completely discouraged. tuberculosis, bleeding disorder, hepatitis, allergy, jaundice,
sexually transmitted disease, malaria (last 6 months),
A replacement blood donor is a friend or a relative
typhoid (last 1 year)?
of the recipient whose donated blood unit is credited to • Have you had recent weight loss, swollen glands, low
the patient (predeposit donation). Blood unit that has grade fever, persistent cough, or repeated diarrhea?
been donated replaces the blood unit used for the patient. • Individuals with multiple sexual partners, homosexuals,
Directed donor donates blood for a specific, named and intravenous drug abusers are likely to be infected
patient and is a friend or a relative of the patient. Patient with AIDS virus. Do you practice any of these?
• Have you taken aspirin in last 3 days? Are you taking
selects his or her own blood donor. Directed donations
any other medications?
can be less safe as the donor may hide significant medical • Have you been vaccinated recently?
information due to pressure of donation. Graft vs. host • Have you had any major surgery or received blood
disease is possible after blood transfusion from a relative. transfusion in the last 6 months?
• Did you have any accidental needle stick (health care
CRITERIA FOR SELECTION OF workers), tattooing, or ear piercing in the last 6 months?
BLOOD DONORS • For women donors: Are you pregnant? Are you breast-
feeding a child? Did you have an abortion in the last 6
Careful selection of blood donors is an essential months?
requirement for safe transfusion practice. It is necessary
to ensure safety of both the donor and the recipient. It is
essential to identify potential health problems for the Box 35.2: Reasons for permanent deferral of a
donor and to prevent transmission of infections through blood donor
transfusion to the recepient. Selection process consists
of obtaining medical history, and performing physical • Positive test for HBV, HCV, HIV
examination and certain laboratory tests. Selection of • Known case of angina pectoris or myocardial infarction
blood donors should be carried out by a qualified • Recipient of clotting factor concentrate
physician or by a person working under his supervision. • High risk behavior for HIV infection
A blood donor questionnaire and consent form needs • Gay or bisexual male
to be filled by the prospective donor before blood • History of viral hepatitis in adult life
• Intravenous drug abuse
donation. Typical questions asked to the prospective
• Recipients of human pituitary-derived growth hormone
donor are shown in Box 35.1. The prospective donor
• Any malignancy, endocrine disease, chronic renal disease,
should be assured that the personal information revealed
chronic liver disease, bleeding disorder, diabetes mellitus
shall be kept confidential. dependent on insulin, asthma
Criteria for selection of blood donors are given below
in short. Reasons for permanent and 1 year deferral are
shown in Boxes 35.2 and 35.3. Donation Interval
To protect the donor from iron deficiency, interval
Age
between two successive donations should be atleast 3
Blood donor should be between 18 and 60 years of age. months.
Collection of Donor Blood, Processing and Storage 343

Box 35.3: Reasons for 1 year deferral


Medications

• History of tattooing, ear-piercing, accidental needle Many donors who are taking drugs are excluded because
stick in health workers of their clinical condition. Other donors are not accepted
• Rabies vaccine or hepatitis B immunoglobulin because the drug they are taking can be potentially
• Rape victims harmful to the recipient, e.g. aspirin (which inhibits
• Sexual contact with a person with hepatitis, AIDS, or platelet function), and drugs with teratogenic action like
with an intravenous drug abuser finasteride, isotretinoin, acitretin, and etretinate, or
• Recipient of blood transfusion or organ transplant
cytotoxic drugs (like cyclophosphamide). Recipients of
human pituitary-derived growth hormone are perma-
Volume of Donation nently debarred due to the risk of Creutzfeldt-Jakob
disease.
A donor weighing 45 kg or more can give 350 ml of blood
along with pilot samples for processing. Dental Treatment

Pregnancy and Lactation Following dental treatment or tooth extraction, a 72-hour


deferral period before donation is required due to the
Pregnant women and lactating mothers (up to 1 year risk of bacteremia.
post-partum) are not accepted.
Skin Piercing
Infectious Diseases
As tattooing, electrolysis, ear piercing, accidental needle
Human Immunodeficiency Virus (HIV) stick in health care workers, or acupuncture carry the
Donors with history of unexplained fever, loss of weight, risk of transmission of hepatitis or HIV infection, blood
lymphadenopathy, and persistent diarrhea should be donation should be deferred for 12 months.
excluded. “High risk” group donors (homosexuals;
Blood Transfusion
intravenous drug abusers; or individuals having contact
with commercial sex workers, with multiple sexual A person should not be accepted as blood donor for 12
partners, or with known acquired immunodeficiency months after receiving blood transfusion or organ
syndrome or HIV positive persons) should be requested transplant.
not to donate blood. Purpose of this policy is elimination
of donors in early (HIV antibody-negative) period of HIV Immunization
infection. There is no deferral for recent immunization with a killed
vaccine, recombinant vaccine, or a toxoid. Nature of
Viral Hepatitis
vaccine received and respective deferral period are as
An individual with history of jaundice within last 1 year follows.
should not be accepted. HBSAg- or anti-HCV positive • Attenuated live virus vaccine for measles, mumps,
subjects are permanently barred from donation. yellow fever, oral polio: 2 weeks.
• Rubella, varicella: 4 weeks.
Malaria • Rabies vaccine following bite by a rabid animal: 1 year
In countries where malaria is endemic, a donor may be • Passive immunization with animal sera: 4 weeks.
accepted after 3 months of asymptomatic period • Hepatitis B immunoglobulin: 1 year.
following malarial attack and after full treatment.
Physical Examination
Illness
• Donor should be in good general health.
Prospective donors with history of diabetes mellitus, • Weight: should be minimum 45 kg.
hypertension, heart disease, renal disease, liver disease, • Blood pressure: systolic blood pressure should be
lung disease, cancer, epilepsy, bleeding disorder, or 100-180 mm of Hg and diastolic 50-100 mm of Hg.
allergic disease are not permitted to donate blood. • Pulse: Pulse rate should be 60-100/min and regular.
344 Essentials of Clinical Pathology

• Temperature: should be normal. 4. Emergency drugs and equipment.


• Donor skin at the venepuncture site should be free of 5. Pilot tubes for collection of blood for testing
scars of needle pricks as they may indicate drug (grouping, cross matching, screening for infectious
addiction. diseases).

Estimation of Hemoglobin Technique


Screening of donors for anemia in blood bank is Blood bag and pilot tubes should be labeled with the
commonly done by copper sulphate specific gravity donor identification number and date of collection before
method. Haemoglobin value should be ≥ 12.5 g/dl. blood collection.
Specific gravity of 1.053 of copper sulphate solution The donor should be under constant observation
is approximately equal to hemoglobin concentration of throughout the procedure of collection. Before blood
12.5 grams/dl. A drop of fingerprick blood is allowed to donation, the prospective donor should have eaten well
fall in this solution from a height of 1 cm. If the drop and taken sufficient fluids. The blood donor lies supine
sinks within 15 seconds time, the hemoglobin level is on a bed. The site for venepuncture is selected in the
acceptable. This method often underestimates the antecubital fossa. A sphygmomanometer cuff is applied
hemoglobin value and leads to unnecessary rejection of to the arm and inflated to 60-70 mm of Hg to make the
donors. In addition, specific gravity of whole blood does veins prominent. The selected area is thoroughly
not depend only on hemoglobin content and is also cleansed with iodine and spirit and allowed to dry.
affected by marked rise in total leukocyte count or plasma The blood collection bag is placed on a weighing
balance that has been kept below the level of the arm. A
protein level; in the presence of these conditions,
loose knot is tied in the tubing close to the venepuncture
hemoglobin will be spuriously normal.
needle.
Phlebotomy is carried out and the blood should start
COLLECTION OF DONOR BLOOD
flowing freely across the tubing into the blood bag. The
In the blood bank, donor blood is collected in a well time of venepuncture is noted in the donor form. The
ventilated, well-lighted, and air-conditioned room. Blood tubing is carefully taped to the needle in place with an
is drawn by qualified physician or by an assistant who adhesive tape.
is well trained and is working under his supervision. The pressure is reduced to 30-40 mm of Hg. The donor
is asked to squeeze a rubber ball or a soft sponge material
Equipments and Materials slowly and continuously to quicken the speed of
donation. The blood and the anticoagulant are mixed
1. Blood bag containing anticoagulant-preservative
gently and periodically in the blood bag. The amount of
solution: Blood from a donor is collected in a closed
blood collected is checked on the weighing balance (1
system of sterile, disposable plastic bag (single,
ml of blood weighs 1.05 gm and the weight of 350 ml of
double, or triple bag depending on component to be
blood is 367 gm). When the weight of the blood bag is as
prepared) with 350 ml capacity. These bags contain
required (367 gm + weight of empty bag and antico-
49 ml of citrate phosphate dextrose adenine (CPDA) agulant), the requisite amount of blood has been
solution. Currently, CPDA-1 solution is commonly collected. The procedure of blood collection should not
used in which blood can be kept stored at 2-6°C for take more than 8 minutes.
35 days. This solution inhibits clotting of blood and The pressure cuff is deflated and the tubing is
provides nutrition for cell metabolism. In India, whole clamped with forceps about 10 cm away from the needle.
blood transfusion is still practiced at many places, and The knot made earlier (close to the needle) is tightened
therefore single bag is commonly used. Double or or a sealing clip is applied.
triple bag systems are used if facilities for separation The tubing is cut between the clamp and the knot/
of components are available. sealing clip. The clamp is removed from the tubing and
2. Sphygmomanometer, weighing scale, blood blood samples (for grouping, cross matching, infectious
weighing balance, sealing clips, artery forceps. disease screening) are collected in appropriate tubes. The
3. Iodine, spirit, sterile cotton swabs, adhesive tape. tubing is then reclamped.
Collection of Donor Blood, Processing and Storage 345

Needle is removed from the vein and pressure is legs are elevated above the level of the head. Cold
applied over the puncture site with sterile cotton gauze. compresses are placed on the neck and forehead. This
The needle is discarded in a special “sharps” container. reaction should be distinguished from hypotensive
Blood remaining in the tubing is non-anticoagulated shock (tachycardia, hypotension, and loss of con-
and is forced back into the blood bag. Bag is inverted sciousness). A severe vasovagal reaction is a contra-
gently several times to mix the blood and the anticoagu- indication for future donations.
lant. Anticoagulated blood is then allowed to run back 2. Hyperventilation: This is seen in first-time donors
into the tubing. The blood bag is placed in the refrigerator who are highly excited. Hyperventilation causes loss
at 4-6°C immediately following collection. If platelet of carbon dioxide. This may result in facial twitching
concentrate is to be prepared, the bag should be kept at or muscular spasms. Relief can be obtained by
room temperature till platelets are separated (within 4 breathing in a paper bag.
hours of collection). 3. Nausea and vomiting
After cessation of bleeding, the venepuncture site is 4. Hematoma at the site of venepuncture
covered with sterile gauze and an adhesive tape. After 5. Infection at venepuncture site or thrombophlebitis
8-10 minutes, the donor is allowed to sit up and guided 6. Inadvertent arterial puncture.
to the refreshment area. The donor is issued a donation Any donor reactions should be recorded on the card
card and is given information about need to drink more issued to the donor.
fluids, activities permissible, and care of venepuncture General procedure after collection of donor blood is
site. outlined in Figure 35.2.

Donor Reactions PROCESSING OF DONOR BLOOD


Donor reactions are rare. This refers to various tests and procedures carried out
1. Syncope or vasovagal attack: This is the most common on the donor blood after collection but prior to cross
reaction. It is due to the action of the autonomic matching. Tests done on donor blood are listed in Table
nervous system and is induced by anxiety, site of 35.1.
blood, or pain. Its features are sweating, slowing of
pulse rate, pallor, coldness of skin, sudden hypo- Changes Occurring during Storage
tension, and sometimes fainting, vomiting, or 1. Loss of viability of red cells: Viability refers to the
incontinence. In such a case, donation is stopped, and capacity of red blood cells to survive in recipient’s

Fig. 35.2: General procedure after collection of donor blood


346 Essentials of Clinical Pathology

Table 35.1: Tests done on donor blood after collection increases oxygen affinity of haemoglobin and reduces
and before cross-matching
release of oxygen to the tissues. 2,3-DPG in red cells
• ABO and Rh grouping tends to return to normal levels within 24 hours of
• Screening and identification of unexpected antibodies transfusion.
• Screening tests for infections transmissible by transfusion 4. Loss of granulocyte function (phagocytic and
– Hepatitis B surface antigen
– Test for antibodies to hepatitis C virus
bactericidal property) occurs within 24 hours and
– Test for antibodies to human immunodeficiency virus loss of platelet function occurs within 48 hours of
– Venereal disease research laboratory (VDRL) test for blood collection. F VIII level declines to 50% by 24
syphilis hours and F V declines to 50% by 10-14 days.
– Blood smear for malaria parasite
5. Decrease in pH of blood
6. Formation of microaggregates: In stored blood,
circulation after transfusion. There is progressive loss
aggregates of aged platelets, leucocytes, and cold
of viability with increasing durtion of storage. Storage
insoluble globulin form. Their number and size
conditions should be such that, after transfusion, at
least 75% of transfused red cells should survive at 24 increase with increasing length of storage.
hours in the recipient’s circulation. Shelf life of the
stored whole blood is based on this criterion. For BIBLIOGRAPHY
whole blood stored in CPDA-1 and maintained at 2°
1. Cheesbrough M. District Laboratory Practice in Tropical
to 6°C, shelf life is 35 days.
Countries. Part 1 and Part 2. Cambridge: Cambridge
2. Depletion of ATP: Progressive loss of ATP with
University Press, 1998.
increasing length of storage causes decreased 2. Henry JB. Clinical Diagnosis and Management by
deformability of red cells and impairment of Na+/ Laboratory Methods. 20th Ed. Philadelphia: WB
K+-ATPase pump. Saunders Company, 2001.
3. Reduction of 2, 3-diphosphoglycerate (2,3-DPG): 3. Kawthalkar SM. Essentials of Haematology. New Delhi:
Progressive depletion of 2, 3-DPG with storage Jaypee Brothers Medical Publishers (P) Ltd, 2006.
36
Screening Tests for Infections
Transmissible by Transfusion

The organisms likely to be transmitted by transfusion • Concealment of relevant medical history by pros-
are usually those, which are prevalent in a particular pective donors
geographic area or population. Organisms transmissible • Specificity and sensitivity of screening tests for
by transfusion are listed in Table 36.1. infections may be poor
According to studies conducted in India, prevalence • Non-implementation of National Transfusion Policy
of transmission of infections through transfusions is • Repeat donor system is non-existent
significantly higher as compared to developed nations. • Collection of donor blood during window period
Some of the reasons include: In India, pre-transfusion testing of donor blood for
• Proportion of replacement and professional dona- agents listed in Table 36.2 is currently mandatory.
tions is high The importance of mandatory testing for infectious
organisms transmissible by transfusion is: (i) some
carriers of disease are often asymptomatic, (ii) some viral
Table 36.1: Microorganisms transmissible by transfusion
infections have long incubation periods, and (iii) it is
• Hepatitis viruses essential to safeguard the health of the recipient.
– Hepatitis A virus (rare) Infections by hepatitis B and C viruses and HIV-1 and
– Hepatitis B virus
HIV-2 are characterized by:
– Hepatitis C virus
• Human immunodeficiency virus (HIV) • Persistence of organisms in circulation for prolonged
– HIV-1 duration without necessarily causing clinical
– HIV-2 manifestations
• Human parvovirus B 19 • Persistence in high titers
• Cytomegalovirus
• Long incubation period
• Epstein Barr virus (rare)
• Human T cell leukemia virus (HTLV) • Ability to cause chronic carrier state
– HTLV-1 • Viability of organisms in blood stored at 4°-6°C.
– HTLV-2
Following principal measures can prevent trans-
Prions mission of infection through transfusion:
• Creutzfeldt-Jakob disease (CJD) and variant CJD • Blood should be collected only from voluntary, non-
Bacteria remunerated donors. All high risk persons (intra-
• Treponema pallidum (syphilis) venous drug abusers, homosexuals, prostitutes, and
• Bacterial contamination of donor unit sexual partners of such persons) and professional
• Brucellosis
donors should be excluded. Standard criteria for
Parasites selection of blood donors should be followed.
• Malaria parasites • All blood donations should be tested for infectious
• Trypanosoma cruzi
agents by screening tests; addition of further
• Toxoplasma gondii
• Babesia microti screening tests like HIV RNA and HCV RNA will
• Leishmania donovani further reduce the risk.
• Universal hepatitis B virus vaccination
348 Essentials of Clinical Pathology

Table 36.2: Mandatory infectious disease testing in blood transfusion practice in India
Disease Test
1. Syphilis Serologic test like Venereal Disease Research Laboratory (VDRL) test
2. Hepatitis B Hepatitis B surface antigen (HBsAg)
3. Human immunodeficiency Anti-HIV 1 and anti-HIV 2 antibodies*
virus (HIV) infection
4. Hepatitis C Anti-HCV antibodies•
5. Malaria Blood smear

* Window periods for HIV and HCV infections are further reduced if tests for HIV RNA and HCV RNA (nucleic acid testing or
NAT) are added. NAT, however, is not currently mandatory in India

• Observation of universal precautions in blood fusion was 6.7%; this was much higher in multi-transfused
collection, processing, storage, and transfusion patients like patients with thalassemia and hemophilia.
• Leukofiltration of blood products Infected hepatocytes release large amounts of hepatitis
• Stringent quality control measures. B surface antigen (HbsAg) into the bloodstream. Presence
of HbsAg indicates active infection. The serological marker
VIRUSES first to appear in HBV infection is HBsAg (as early as 5
days after infection).
Hepatitis B Virus (HBV)
Screening of all blood donations for HbsAg has
HBV is a partially double-stranded DNA virus of 42 nm greatly reduced the risk of transmission of HBV through
diameter (Fig. 36.1). It can cause acute hepatitis, chronic transfusion.
hepatitis, asymptomatic carrier state, cirrhosis, or Tests for screening donor blood for HbsAg are:
hepatocellular carcinoma. • Reverse passive hemagglutination assay (RPHA)
In India, prevalence of HBsAg carriers is reported to • Enzyme linked immunosorbent assay (ELISA)
be 1.5 to 4%. HBV is highly infectious and is transmitted • Radioimmunoassay (RIA).
through all blood components and most of the blood Commercial test kits for detection of HbsAg are
derivatives. According to one study in India, incidence available and the exact test procedure is provided with
of post-transfusion hepatitis following a single trans- each kit. General principles of these tests are outlined
below.

Reverse Passive Hemagglutination Assay


In RPHA, red cells that are coated with anti-HBs antibody
are added to the donor’s serum. If HbsAg is present in
the serum, agglutination of red cells will occur. Absence
of agglutination indicates negative test. The test is called
as ‘passive reverse agglutination’ because antibody, and
not antigen, is artificially coated on to the red cells. The
test is performed in U- or V-shaped wells of a microtiter
plate which can be centrifuged to augment agglutination.
The test is sensitive to 10-100 ng of HbsAg antigen per
ml of serum (Fig. 36.2).

Enzyme-linked Immunosorbent Assay

Fig. 36.1: Diagrammatic representation of Serum sample to be tested is incubated with anti-HbsAg
hepatitis B virus antibody which has been coated to microtitre plate. The
Screening Tests for Infections Transmissible by Transfusion 349

solution (an acid) is added to prevent any further reaction


between the enzyme and the substrate. The result is read
in a spectrophotometer at the specified wavelength.

Hepatitis C Virus (HCV)


HCV is a single-stranded RNA virus of flaviviridae
family. Incubation period is about 8 weeks. Most cases
of HCV infection are asymptomatic. Some patients
develop chronic hepatitis, cirrhosis, or hepatocellular
carcinoma.
In India, prevalence of HCV is reported to be 1.66%.
As the amount of HCV antigen in bloodstream is
small and cannot be detected readily, screening of donor
blood for HCV infection is done by detection of anti-HCV
antibody in serum (becomes detectable after 6-8 weeks
of infection).
The earlier tests (first generation ELISA) used
recombinant proteins complementary to the NS4 region
Fig. 36.2: Principle of reverse passive hemagglutination
assay for HBsAg (c100-3) of the HCV genome. Second generation ELISA
incorporated recombinant or synthetic antigens from
NS4 as well as NS3 regions (c100-3, c22-3, c33c) of the
test serum is incubated in the well during which binding genome, resulting in improvement in sensitivity and
of antigen from serum (if present) and antibody (attached specificity. The third generation ELISA, in addition to
to the well) occurs. An enzyme-linked anti-HBs antibody NS3 and NS4, also includes antigens from NS5 region
(conjugate) is added which will combine with the bound (Fig. 36.3).
antigen. A chromogenic enzyme substrate is finally
added and the mixture is incubated in the dark. If enzyme Human Immunodeficiency Virus
is present (bound to the antigen), then its action on the Human immunodeficiency virus or HIV is a RNA
substrate will lead to the colour development. A stopping retrovirus (Fig. 36.4), which causes slowly progressive

Fig. 36.3: Hepatitis C virus genome and the recombinant proteins


350 Essentials of Clinical Pathology

Fig. 36.4: Diagrammatic representation of human


immunodeficiency virus Fig. 36.5: Principle of ELISA test for anti-HIV antibodies

immunodeficiency in infected persons. The cells most ELISA, and (ii) HIV-1 nucleic acid amplification testing
susceptible to HIV infection are CD4+ T lymphocytes. (NAT). NAT testing is based on polymerase chain reaction.
Destruction of CD4+ T lymphocytes results in slowly
progressive impairment of the immune system. The
BACTERIA
infected individual becomes susceptible to a range of Treponema pallidum
opportunistic infections and malignancies. The most Blood transfusion can transmit Treponema pallidum, the
advanced stage of HIV disease is acquired immune causative agent for syphilis. It is a spirochete that can be
deficiency syndrome or AIDS. visualized by dark ground illumination (Fig. 36.6). T.
According to the estimates of India’s National AIDS pallidum is destroyed by storage of blood at 2-8°C for 48-
Control Organization (NACO), adult prevalence of HIV 72 hours. However, fresh blood or platelet concentrates
infection is 0.7% with approximately 4 million HIV can transmit these organisms. Transfusion-transmission
infections, 90% of which are in the age group of 15-45 of syphilis is, therefore, rare. The main value of testing
years. donor blood for T pallidum is to identify and exclude
donors with high-risk behavior and thus who are at risk
Following HIV infection, viremia becomes detectable
of having sexually transmitted infections.
after a few days and lasts for several weeks. Anti-HIV
antibodies appear 6-12 weeks after infection (called as
seroconversion). Principle of ELISA test for detection of
anti-HIV antibodies is shown in Figure 36.5. Window
period is the period between the onset of HIV infection
and appearance of detectable anti-HIV antibodies in
serum; it is the infectious but seronegative period (i.e.
the test for anti-HIV antibodies is yet to become positive).
Transfusion of donor blood collected during the window
period will transmit HIV to the recipient.
According to American Association of Blood Bank
standards, HIV screening tests necessary for whole blood Fig. 36.6: Treponema pallidum observed under dark
donors are (i) Anti-HIV antibodies (HIV-1 and HIV-2) by ground illumination
Screening Tests for Infections Transmissible by Transfusion 351

Screening of donor blood for antibody to T. pallidum is


usually carried out by rapid plasma reagin test or Venereal
Disease Research Laboratory (VDRL) test. In VDRL test
that is a non-specific test, donor serum (heated to 56°C to
inactivate the complement) is mixed with cardiolipin-
lecithin-cholesterol antigen. If flocculation is observed, the
test is reported as reactive (Fig. 36.7).

PARASITES

Plasmodium Species
Malaria parasite can be transmitted through all blood
components. For detection of malaria parasite, commonly
blood smears are examined by light microscopy.
However, the test is positive only when parasites are
> 100/μl in blood.

BIBLIOGRAPHY
1. Chatterjee K, Sen A. Step by Step Blood Transfusion
Services. A Practical Manual on the Technical and
Clinical Aspects. New Delhi: Jaypee Brothers and
Medical Publishers (P) Ltd, 2006.
Fig. 36.7: Principle of Venereal Disease Research 2. Kawthalkar SM. Essentials of Haematology. New Delhi:
Laboratory (VDRL) slide test for syphilis Jaypee Brothers Medical Publishers (P) Ltd, 2006.
37

Compatibility Test
(Cross-match)

When the recipient’s ABO and Rh blood groups are


determined, the donor blood unit that is ABO and Rh
compatible is selected, and compatibility test is carried
out. The purpose of compatibility test is to prevent the
transfusion of incompatible red cell units and thus
avoidance of hemolytic transfusion reaction in the
recipient. Compatibility test detects (i) major ABO
grouping error, and (ii) most clinically significant
antibodies reactive against donor red cells.
There are two types of cross-match: major cross-match
(testing recipient’s serum against donor’s red cells) and
minor cross-match (testing donor’s serum against
recipient’s red cells). However, minor cross-match is
considered as less important since antibodies in donor
blood unit get diluted or neutralized in recipient’s
plasma. Also, if antibody screening and identification is
being carried out, minor cross-matching is not essential.
Therefore, only the red cells from the donor unit are
tested against the recipient’s serum and the name Fig 37.1: Immediate spin cross-match
compatibility test has replaced the term cross-matching.
For transfusion of platelets or fresh frozen plasma,
cross-matching is not required. However, fresh frozen
plasma should be ABO-compatible.
Causes of False-negative Test
A full cross-matching procedure consists of:
• Immediate spin cross-match at room temperature, and 1. A2B donor red cells and group B recipient serum.
• Indirect antiglobulin test at 37°C. 2. Rapid complement fixation of potent ABO antibodies
with bound complement interfering with aggluti-
IMMEDIATE SPIN CROSS-MATCH nation.
• The purpose of this test is to detect ABO incompati-
Causes of False-positive Test
bility. Equal volumes of 2% saline suspension of red
cells of donor and recipient’s serum are mixed, 1. Rouleaux formation
incubated at room temperature for 5 minutes, and 2. Cold-reactive antibodies: If agglutination disappears
centrifuged. Agglutination or hemolysis indicates by keeping the tube at 37°C for 10 minutes, presence
incompatibility (Fig. 37.1). of cold agglutinins is confirmed.
Compatibility Test (Cross-match) 353

recipient’s serum. Agglutination or hemolysis at any stage


is indicative of incompatibility.

EMERGENCY CROSS-MATCH
If blood is required urgently, ABO and Rh grouping are
carried out by rapid slide test and immediate spin cross
match (i.e. the first stage of cross match) is performed
(to exclude ABO incompatibility). If the blood unit is
compatible, then after issuing it, remaining stage of the
cross-match is completed. If any incompatibility is
detected, the concerned physician is immediately
informed about the incompatibility detected.

ANTIBODY SCREENING AND


IDENTIFICATION
Screening for unexpected or irregular antibodies is done
during pre-transfusion testing in recipient’s serum and
Fig 37.2: Indirect antiglobulin test for detection of in donor’s blood. In this test, serum of the recipient is
clinically significant IgG antibodies tested against a set of three group O screening cells of
known antigenic type. If unexpected antibodies are
INDIRECT ANTIGLOBULIN TEST detected, then they are identified and blood unit that
lacks the corresponding antigen is selected for compati-
Saline-suspended red cells of the donor after being
bility test.
incubated in patient’s serum are washed in saline and
antiglobulin reagent is added. Following re-centri-
BIBLIOGRAPHY
fugation, examine for agglutination or hemolysis
(Fig. 37.2). This test detects most of the clinically 1. Kawthalkar SM. Essentials of Hematology. New Delhi:
Jaypee Brothers Medical Publishers (P) Ltd, 2006.
significant IgG antibodies. 2. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical
If agglutination or hemolysis is not observed in any Hematology (9th Ed). London: Churchill Livingstone,
of the above stages, donor unit is compatible with 2001.
38

Adverse Effects of
Transfusion

Blood transfusion is a life saving procedure in an the recipient. It results from transfusion of ABO-
appropriate setting and there are no side-effects in mismatched blood to the recipient due most commonly
majority of cases. However, it is a potentially harmful to a clerical error. Most severe reaction occurs if group A
procedure and every recipient of transfusion is at risk of blood is transfused to a group O recipient.
an adverse reaction. It should be prescribed only if there Pathophysiology consists of antigen-antibody
is a definite clinical indication. This is because even with reaction that leads to complement activation and
best possible blood banking standards, transmission of intravascular hemolysis. This causes hypotension, shock,
infections or other complications can occur. acute renal failure, and disseminated intravascular
Adverse effects of transfusion are listed in Table 38.1. coagulation (Fig. 38.1). Signs and symptoms (that appear
Main causes of transfusion-related deaths are: within minutes of starting transfusion) include fever,
• Immediate acute hemolytic transfusion reaction (ABO pain at the infusion site, loin pain, tachycardia,
incompatibility) hemoglobinuria, and hypotension. In anesthetized
• Pulmonary edema and congestive heart failure patients, bleeding and hypotension are the only
(circulatory overload) indications.
• Bacterial contamination of blood unit
• Transfusion of physically damaged red cells (e.g. by
heat, cold)
• Transfusion-associated graft vs. host disease

ACUTE HEMOLYTIC TRANSFUSION


REACTION
This is a medical emergency and results from intra-
vascular destruction of donor red cells by antibodies in

Table 38.1: Adverse effects of transfusion


Immediate Delayed
1. Acute hemolytic 1. Delayed hemolytic
transfusion reaction transfusion reaction
2. Febrile non-hemolytic 2. Transmission of infections
transfusion reaction
3. Allergic reactions 3. Iron overload
4. Anaphylactic reactions 4. Graft vs. host disease
5. Transfusion-associated 5. Post-transfusion purpura
lung injury
6. Volume overload
7. Bacterial contamination
of donor unit Fig. 38.1: Pathophysiology of acute hemolytic
transfusion reaction
Adverse Effects of Transfusion 355

Laboratory features are: Gram staining and culture of blood from the blood bag
• Hemoglobinemia (pink coloration of plasma after and from the recipient. Direct antiglobuin test is negative.
centrifugation of post-transfusion sample)
• Positive direct antiglobulin test TRANSFUSION-ASSOCIATED LUNG INJURY
• Hemoglobinuria This is an acute respiratory disorder that manifests with
• Schistocytes (fragmented red cells) and spherocytes fever, chills, dyspnea, and dry cough. X-ray shows diffuse
on blood smear pulmonary infiltrates. One probable mechanism is
• Elevated indirect serum bilirubin reaction of anti-HLA or anti-neutrophil antibodies in
donor blood with leukocytes of the recipient leading to
FEBRILE NON-HEMOLYTIC the formation of leukocyte aggregates; these aggregates
TRANSFUSION REACTION deposit in pulmonary vasculature and cause increased
This is the most common transfusion reaction. It occurs vascular permeability and pulmonary edema.
in about 1% of all transfusions and is defined as an
unexplained rise of temperature of atleast 1°C during or DELAYED HEMOLYTIC
shortly after transfusion. It is caused by the release of TRANSFUSION REACTION
pyrogenic cytokines from white cells (during storage of This is a hemolytic transfusion reaction occurring several
blood unit or following transfusion due to reaction of days or weeks after transfusion. This occurs in indi-
alloantibodies with white cells of donor). This reaction viduals who have been sensitized to a red cell antigen
is common in multiply-transfused patients. Signs and by a previous transfusion or pregnancy so that the
symptoms include fever, chills, and tachycardia. antibody is present in a low titer. On re-exposure, there
Diagnosis depends on exclusion of other causes of febrile is a secondary IgG immune response and mainly
transfusion reaction. exravascular hemolysis. This reaction is typically
Transfusion reactions presenting with fever are associated with Kidd antibodies.
shown in Figure 38.2. Signs and symptoms include fever, mild jaundice,
and mild anemia. Laboratory features include raised
BACTERIAL CONTAMINATION OF indirect serum bilirubin, spherocytes on blood smear,
DONOR UNIT anemia, and positive direct antiglobulin test.
Transfusion of an infected blood product is more Acute and delayed hemolytic transfusion reactions
common with platelet concentrates since platelets are are compared in Table 38.2.
stored at a higher temperature (20-24°C) that promotes
multiplication of contaminating bacteria. Organisms ANAPHYLACTIC REACTION
depend on the nature of blood product. Platelets are This rare reaction occurs in IgA-deficient recipients in
usually contaminated with gram-positive cocci, while red whom anti-IgA antibodies react with IgA in donor
cells are contaminated with Yersinia enterocolitica, plasma, leading to activation of complement and
Escherichia coli, or Pseudomonas species. formation of anaphylatoxins (C3a and C5a). Signs and
Signs and symptoms include high grade fever with symptoms include development of acute hypotension,
rigors, hypotension, and shock. Laboratory studies shock, and dyspnoea after transfusion of a few drops of
include inspection of blood bag for discoloration, and blood.

Fig. 38.2: Transfusion reactions presenting with fever


356 Essentials of Clinical Pathology

Table 38.2: Comparison of acute and delayed hemolytic transfusion reactions


Parameter Acute Delayed
1. Type of antibody Anti-ABO Anti-Kidd
2. Nature of antibody Naturally-occurring Immune
3. Time of appearance of Within minutes After several days or weeks
clinical features after
transfusion
4. Site of hemolysis Intravascular Extravascular
5. Clinical features Fever, chills, back pain, Fever, mild jaundice, anemia
acute renal failure, DIC

ALLERGIC REACTION antigen (anti-HBc) may be the only evidence of disease.


This results from type I hypersensitivity reaction to some HBsAg-positive donors are permanently excluded from
donor plasma proteins. It is the second most frequently donations.
reported transfusion reaction. Signs and symptoms Liver diseases caused by hepatitis B virus include:
include mild urticaria, rash, and pruritus. subclinical hepatitis, acute icteric hepatitis, fulminant
hepatitis (massive hepatic necrosis), chronic hepatitis,
VOLUME OVERLOAD cirrhosis, and hepatocellular carcinoma.
3. Hepatitis C virus: In India, prevalence of hepatitis C
This occurs if transfusion rate is too rapid, or excessive,
virus is reported to be 1.66% and there are 15 million
or if cardiac or renal impairment is present. It causes
carriers. This RNA virus is the most common cause
cardiac failure and lung edema.
of transfusion-transmitted hepatitis. It is transmitted
IRON OVERLOAD by both cellular and plasma components. Incubation
period is about 8 weeks. Persistent infection is the
Each unit of blood contains 200 mg of iron. Patients rule. Chronic hepatitis is very common (usually mild).
receiving regular transfusion therapy such as those with Cirrhosis occurs in a minority of patients, and
thalassemia develop manifestations of parenchymal hepatocellular carcinoma occurs in about 10% with
damage due to iron accumulation. cirrhosis.
The test used for donor screening is anti-HCV
TRANSMISSION OF INFECTIONS antibody test. From infection till the appearance of
Organisms transmissible by transfusion are listed in anti-HCV positivity (70-80 days), HCV RNA can be
Table 36.1 (See Chapter 36: Screening Tests for Infections detected by PCR testing.
Transmissible by Transfusion). 4. Human immunodeficiency virus (HIV): In India,
1. Hepatitis A virus: Hepatitis A is rarely transmitted adult prevalence is 0.7%. Two genetically different
by transfusion, as the duration of viremia is short. but closely related viruses are recognized: HIV-1 and
Donors with hepatitis A or who are in close contact HIV-2. Both are RNA retroviruses. Natural history
with a hepatitis A are deferred for 1 year. of HIV infection consists of (i) acute, self-limited, “flu-
2. Hepatitis B virus: In India, prevalence of hepatitis B like” illness occurring 3-6 weeks after infection, (ii)
virus is 1.5 to 4%, and there are 43 million carriers. chronic phase lasting for several years that may be
Hepatitis B virus, a DNA virus, can be transmitted asymptomatic or associated with persistent lymph-
by both cellular and plasma components. Incubation adenopathy, and (iii) final, full-blown phase with
period is 2-6 months. All blood donations are tested opportunistic infections and malignancies. HIV can
for HBsAg by a sensitive method that has greatly be transmitted by both cellular and plasma compo-
reduced the risk of transmission. During early period nents.
of infection, HBsAg may be undetectable; during this The test used for detection of infection is anti-
window period, antibodies to the hepatitis B core HIV-1/2 antibodies by enzyme immunoassay. To
Adverse Effects of Transfusion 357

reduce the window period from infection to appea- form gradually in stored blood. Rapid transfusion of large
rance of antibodies from about 22 days to 10 days, volumes of stored blood leads to:
nucleic acid testing (NAT) for HIV RNA is • Dilution of platelets and coagulation factors
recommended. • Hyperkalemia (due to release of potassium from
5. Treponema pallidum: Trasfusion-transmission of stored red cells)
syphilis is rare since T. pallidum does not survive in • Hypocalcemia (due to binding of calcium by citrate)
refrigerated storage and is inactivated at 4°C after 4 • Hypothermia (due to rapid infusion of large amount
days; however, fresh blood and platelet concentrates of cold blood)
can transmit the organism. The main value of • Adult respiratory distress syndrome due to migration
screening test is as a marker of high-risk behavior. of microaggregates to lungs.
6. Malaria parasites: Malaria parasites are readily RECOGNITION AND INVESTIGATION OF A
transmitted by transfusion. In endemic areas, it is not TRANSFUSION REACTION
practical to reject all potential donors with history of
All reactions following blood transfusion should be
malaria in the past. In endemic areas, the only safe
considered as hemolytic in nature and should be
prevention is administration of preventive anti-
investigated accordingly (Fig. 38.3).
malarial drugs to all recipients of transfusion.
1. Transfusion should be immediately stopped, leaving
open intravenous line with normal saline.
COMPLICATIONS ASSOCIATED WITH 2. All paperwork and blood bag should be checked for
MASSIVE TRANSFUSION clerical error. More than 90% of hemolytic trans-
fusions result from a clerical error (i.e. a wrong unit
Massive transfusion refers to transfusion of stored blood of blood is given to the wrong recipient).
equivalent to patient’s blood volume in 24 hours. 3. Blood bank is informed immediately and the blood
Morbidity and mortality is due to rapid blood loss bag, administration set, and post-transfusion blood
coupled with transfusion of stored blood. and urine samples should be sent to the blood bank.
Storage of blood is associated with loss of 2,3- 4. Evidence of hemolysis: Obtain a post-transfusion
diphosphoglycerate, lowering of pH, loss of ATP, loss of blood sample from the recipient, centrifuge, and
platelet function, and depletion of coagulation factors. observe for pink discoloration of overlying plasma
Microaggregates composed of leukocytes and platelets (hemoglobinemia); this is the most rapid way of

Fig. 38.3: Immediate management of a suspected transfusion reaction. All reactions should be assumed to be
hemolytic and investigated accordingly
358 Essentials of Clinical Pathology

detecting intravascular hemolysis if pre-transfusion 6. Investigations for detection of complications of


sample is normal. Similarly, visual examination of hemolytic transfusion reaction:
patient’s urine can be done for hemoglobinuria. • Tests for disseminated intravascular coagulation:
Blood smear examination will show fragmented red Blood smear, coagulation screen, and test for fibrin
cells and spherocytes. Indirect serum bilirubin is
degradation products
raised.
• Tests for acute renal failure: Blood urea, serum
5. Evidence of blood group incompatibility: Perform
a direct antiglobulin test (DAT) on post- and pre- creatinine, and serum electrolytes
transfusion blood samples. Positive DAT on post- 7. Bacteriological culture if the cause of the acute
transfusion sample (with negative test on pre- transfusion is still not clear.
transfusion sample) is indicative of an immunological
hemolytic transfusion reaction. Blood group incom- BIBLIOGRAPHY
patibility will also be revealed on (i) repeat ABO
1. Kawthalkar SM. Essentials of Hematology. New Delhi:
grouping on recipient’s pre- and post-transfusion Jaypee Brothers Medical Publishers (P) Ltd, 2006.
samples and on donor unit, and (ii) repeat cross- 2. World Health Organization: Blood Transfusion Safety:
matching of donor blood against recipient’s pre- and The Clinical Use of Blood. Geneva: World Health
post-transfusion samples. Organization, 2002.
39

Blood Components

A single whole blood donation can be separated into gation due to differences in their specific gravities. After
different components to provide treatment to more than their separation, various components can be
one patient. One unit of whole blood can be broken down transferred from one bag to another in a closed circuit
into one unit of packed red cells, one unit of platelets, thus avoiding exposure to external environment and
and one unit of fresh frozen plasma/cryoprecipitate. This maintaining the sterility. Blood should be processed
practice avoids wastage of collected whole blood (each for component separation within 6 hours of collection
component is stored at a temperature that is optimal for (Figs 39.1 and 39.2).
that component), allows administration of specific 2. Apheresis: This is a procedure in which a suitable donor
replacement therapy, and also avoids transfusion of is connected to an automated cell separator machine
unnecessary blood elements that are not required by the (that is essentially designed as a centrifuge) through
which whole blood is withdrawn, the desired blood
patient.
component is retained, and the remainder of the
Terms used in transfusion therapy are shown in Table
blood is returned back to the donor. Depending on
39.1.
the component that is separated and removed, the
There are two methods for collection of blood for
procedure is called as plateletpheresis, leukapheresis,
preparation of blood components:
or plasmapheresis.
1. Single whole blood donation: Preparation of blood
components has been greatly facilitated by the
WHOLE BLOOD
introduction of double and triple bags having closed
integral tubing. After collection of a unit of whole Whole blood is one unit of donor blood collected in a
blood in the primary bag, blood components can be suitable anticoagulant-preservative solution (citrate
separated from one another by differential centrifu- phosphate dextrose adenine or CPDA-1). Its total volume

Table 39.1: Definitions used in transfusion therapy

Blood product
A therapeutic substance prepared from human blood
Whole blood
One unit of non-separated donor blood collected in an appropriate container containing anticoagulant-preservative
solution
Blood component
A constituent separated from whole blood by differential centrifugation or that is obtained directly from donor by apheresis
Plasma derivative
Human plasma proteins obtained from multiple donor units of plasma under pharmaceutical manufacturing conditions.
These products are heat-treated or chemical-treated to inactivate lipid-enveloped viruses.

Plasma derivatives like factor concentrates and immunoglobulins can also be prepared by recombinant DNA technology
360 Essentials of Clinical Pathology

Fig. 39.1: Blood components are prepared from a unit of whole blood within 6 hours of collection. Initial light centrifugation
separates red cells from platelets and plasma. Heavy centrifugation of platelet-rich plasma separates platelets and plasma

Fig. 39.2: Principle of preparation of blood components from one unit of whole blood

is about 400 ml (350 ml of blood + 49 ml of anticoagulant). F VIII). Transfusion of whole blood should commence
It consists of cellular elements and plasma. Whole blood within 30 minutes of removal from the refrigerator, and
is stored in an approved blood bank refrigerator at should be complete within 4 hours of starting. Trans-
fusion of one unit raises hemoglobin by 1 gm/dl or
4°-6°C. Shelf life of such blood (collected in CPDA hematocrit by 3%.
anticoagulant) is 35 days. It does not contain functionally Indications and contraindications for whole blood
effective platelets and labile coagulation factors (F V and transfusion are given in Table 39.2.
Blood Components 361

Table 39.2: Indications and contraindications for whole Table 39.4: Indications for packed red cell transfusion
blood transfusion
• Anemia: Chronic severe anemia, severe anemia with
Indications congestive cardiac failure, anemia in elderly
• Acute blood loss with hypovolemia • Acute blood loss (transfused along with a crystalloid
• Exchange transfusion in neonates or a colloid solution)
• Non-availability of red cell concentrate or suspension

Contraindications blood in the primary collection bag (containing CPDA-


• Chronic anemia with compromised cardiovascular 1), maximum amount of plasma is removed (after
function centrifugation) and transferred to one satellite bag. The
additive solution from the second satellite bag is
transferred into the primary collection bag (containing
BLOOD COMPONENTS packed red cells) in a closed system.
Blood components are listed in Table 39.3. Indications for red cells in SAG-M are similar to
those for packed red cells.
RED CELL COMPONENTS 3. Leukocyte-poor red cells: Leukocyte-poor red cells
contain < 5 × 106 white cells per bag. Methods for
1. Packed red cells: Packed red cells are prepared by leukocyte depletion are (i) leukocyte-reduction filters,
removing most of the plasma from one unit of whole and (ii) removal of buffy coat. Indications for
blood (hematocrit 70-75%). Whole blood is either leucocyte-poor red cells are: (i) prevention of HLA
allowed to sediment overnight in a refrigerator at 2- immunization in patients who are likely to receive
6°C or is spun in a refrigerated centrifuge. Super- allogeneic bone marrow transplantation, (ii) preven-
natant plasma is then separated from red cells in a tion of febrile nonhemolytic transfusion reactions in
closed system by transferring it to the attached empty persons receiving multiple transfusions, and (iii)
satellite bag. Red cells and a small amount of plasma prevention of transmission of cytomegalovirus.
are left behind in the primary blood bag. Packed red 4. Washed red cells: Red cells can be washed with
cells have a high viscosity and therefore the rate of normal saline to remove plasma proteins, white cells,
infusion is slow. Transfusion of one unit of red cells and platelets. Such red cells are used for IgA-deficient
increases hemoglobin by 1 gm% (or increases individuals who have developed anti-IgA antibodies,
hematocrit by 3%). as exposure will lead to anaphylaxis.
Indications for packed red cells are shown in 5. Frozen red cells: If a cryoprotective agent such as
Table 39.4. glycerol is added, red cells can be stored frozen for
2. Red cells in additive solution (Red cell suspension): upto 10 years. This method can be used for storage
These are red cells with minimal residual plasma and for donor red cells with rare blood groups, for future
an additive solution (SAG-M which contains saline, autologous transfusion, and for individuals who have
adenine, glucose, and mannitol). This increases shelf repeated febrile nonhemolytic transfusion reactions.
life from 35 days to 42 days. After collection of whole 6. Irradiated red cells: Gamma-irradiation of red cells
inactivates lymphocytes and prevents graft vs. host
Table 39.3: Blood components disease. Irradiated red cells are indicated for
intrauterine or premature neonate transfusions, and
Cellular components
in individuals with immunodeficiency, and in those
• Red cells: Packed red cells, red cells in additive
solution, leukocyte-poor red cells, washed red cells, receiving blood from first-degree relative donors.
frozen red cells, irradiated red cells
• Platelets: Platelet concentrate, apheresis platelets
PLATELETS
• Granulocytes: granulocyte concentrate Platelet concentrates can be obtained from single donor
units or by plateletpheresis.
Plasma components
1. Platelet concentrate (Random donor platelets prepared from
• Fresh frozen plasma
whole blood unit): One unit of whole blood is
• Cryoprecipitate
centrifuged (light spin) to obtain platelet-rich plasma
362 Essentials of Clinical Pathology

(PRP). PRP is then transferred to the attached satellite Table 39.6: Indications for fresh frozen plasma
bag and spun (high spin) to get platelets at the bottom • Multiple coagulation factor deficiencies: liver disease,
and supernatant plasma. Most of the supernatant is warfarin overdose, massive blood transfusion
returned back to the primary collection bag or to • Disseminated intravascular coagulation
another satellite bag, leaving behind 50-60 ml of • Inherited deficiency of a coagulation factor for which
plasma with the platelets. no specific replacement therapy is available
Platelets are stored at 20°-24°C with continuous • Thrombotic thrombocytopenic purpura
agitation (in a storage device called platelet agitator).
Maximum period of storage is 5 days. separated from whole blood by centrifugation,
One unit of platelet concentrate contains > 45 × expressed into the attached satellite bag, and rapidly
109 platelets. Transfusion of one unit will raise the frozen at –20°C or at lower temperature. FFP contains
platelet count in the recipient by about 5000/μl. all the coagulation factors.
The usual adult dose is 4-6 units of platelet FFP can be stored for 1 year if temperature is
concentrate (or 1 unit/10 kg of body weight). These maintained below –25°C. When required for transfusion,
units (which are from different donors) are pooled FFP is thawed between 30-37°C and then stored in the
into one bag before transfusion. This dose will raise refrigerator at 2-6°C. Since labile coagulation factors
the platelet count by 20,000 to 40,000/μl. rapidly deteriorate, FFP should be transfused within 2
2. Plateletpheresis (Single donor platelets): In platelet hours of thawing.
pheresis, a donor is connected to a blood cell separator Indications for FFP are shown in Table 39.6.
machine in which whole blood is collected in an 2. Cryoprecipitate: Cryoprecipitate is prepared from
anticoagulant solution, platelets are separated and plasma that has been freshly separated (within 6
retained, and remaining components are returned hours of collection) by rapidly freezing it at -20°C or
back to the donor. With this method, a large number lower and thawing it slowly at 4-6°C. A white
of platelets can be obtained from a single donor flocculent precipitate and plasma are obtained. The
(equivalent to 6 units of platelet concentrate). This mixture is centrifuged and supernatant plasma is
method is especially suitable if HLA-matched removed leaving behind sediment of cryoprecipitate
platelets are required (i.e. if patient has developed suspended in 10-20 ml of plasma. The unit is then
refractoriness to platelet transfusion due to the refrozen (-20°C or colder) and can be stored at this
formation of alloantibodies against HLA antigens). temperature for 1 year. When needed, cryoprecipitate
The usual indications and contraindications for is thawed at 30-37°C, required donations are pooled
administering platelets are shown in Table 39.5. and transfused to the patient. Cryoprecipitate
contains F VIII, von Willebrand factor, fibrinogen, F
PLASMA COMPONENTS
XIII, and fibronectin. Indications for cryoprecipitate
The main plasma components are fresh frozen plasma are F VIII deficiency (if F VIII concentrate is not
and cryoprecipitate. available), von Willebrand disease, and deficiency of
1. Fresh frozen plasma (FFP): FFP is prepared from fibrinogen.
whole blood within 6 hours of collection because after
this time labile coagulation factors are lost. Plasma is
PLASMA DERIVATIVES
Table 39.5: Indications and contraindications to Plasma derivatives are manufactured by fractionation of
platelet transfusions large volumes of pooled human plasma. Important
Indications plasma derivatives are listed in Table 39.7.
• Bleeding due to decreased platelet production 1. Human albumin solutions: Albumin is prepared by
• Bleeding in hereditary disorders of platelet function cold ethanol fractionation of pooled plasma and is
• Massive blood transfusion
sterilized during manufacture to destroy viruses and
Contraindications bacteria. Albumin is used as a replacement fluid in
• Thrombotic thrombocytopenic purpura therapeutic plasma exchange, and for treatment of
• Hemolytic uremic syndrome
diuretic-resistant edema of hypoproteinemia.

tahir99 - UnitedVRG
vip.persianss.ir
Blood Components 363

Table 39.7: Plasma derivatives a. Non-specific immunoglobulins: These are derived


1. Human albumin solutions from the pooled plasma of non-selected donors.
2. F VIII concentrate Some indications include (i) passive prophylaxis
3. F IX concentrate of viral infections like hepatitis, rubella, and
4. Prothrombin complex concentrate measles, (ii) treatment of hypogamma-
5. Immunoglobulins globulinaemia, (iii) autoimmune thrombocyto-
paenic purpura to induce a rise in platelet count,
2. F VIII concentrate: Freeze-dried F VIII concentrate is and (iv) neonatal sepsis.
prepared by fractionation from large pools of fresh b. Specific immunoglobulins: They are obtained from
frozen plasma. To reduce the risk of transmission of donors who have selected high titer IgG antibodies.
viral infections, it is treated with heat or chemicals Anti-RhD immunoglobulin is prepared from
during manufacturing process. F VIII concentrate is plasma of Rh-negative donors who have produced
the treatment of choice for treatment of hemophilia anti-D following immunization; it is used for
A and severe von Willebrand disease. prevention of sensitization to RhD antigen in Rh-
3. Prothrombin complex concentrate (PCC): PCC negative women giving birth to a Rh-positive baby.
contains factors II, VII, IX, and X, and also protein C Other specific immunoglobulins include hepatitis
and S. Main uses of PCC are (i) deficiency of F IX, (ii) B immune globulin, varicella-zoster immune
deficiency of F VIII with development of inhibitors globulin, and tetanus immune globulin that are
against F VIII, and (iii) inherited deficiency of factors used for passive prophylaxis of infections.
II, VII, and X. A serious risk of PCC is thrombotic
complications due to the presence of small amounts BIBLIOGRAPHY
of activated coagulation factors.
1. Kawthalkar SM. Essentials of Hematology. New Delhi:
4. Immunoglobulins: Immunoglobulins are obtained Jaypee Brothers Medical Publishers (P) Ltd, 2006.
by cold ethanol fractionation of large pools of human 2. World Health Organization. Blood Transfusion Safety:
plasma. They are of two types: specific and non- The Clinical Use of Blood. World Health Organization.
specific. Geneva, 2002.

tahir99 - UnitedVRG
vip.persianss.ir
General References

1. Cheesbrough M. District laboratory practice in tropical countries. Part 1 and Part 2. Cambridge. Cambridge University
Press, 1998.
2. Crook MA. Clinical chemistry and metabolic medicine (7th Ed). London: Edward Arnold (Publishers) Ltd, 2006.
3. Wallach J. Interpretation of diagnostic tests (7th Ed). Philadelphia: Lippincott Williams and Wilkins, 2000.
4. Mitchell RN, Kumar V, Abbas AK, Fausto N. Robbins and Cotran pathologic basis of disease (7th Ed). Philadelphia:
Saunders, 2006.
5. Henry JB. Clinical diagnosis and management by laboratory methods (20th Ed). Philadelphia: WB Saunders Company,
2001.
6. Burtis CA, Ashwood ER. Tietz fundamentals of clinical chemistry (5th Ed). Philadelphia: WB Saunders Company,. 2001.
7. World Health Organization. Manual of basic techniques for a health laboratory (2nd Ed). Geneva: World Health
Organization, 2003.
8. Provan D, Krentz A. Oxford Handbook of Clinical and Laboratory Investigation. Oxford. Oxford University Press, 2002.
9. Lewis SM, Bain BJ, Bates I. Dacie and Lewis Practical Haematology (9th Ed). London: Churchill Livingstone, 2001.
10. Provan D, Singer CRJ, Baglin T, Lilleyman J. Oxford Handbook of Clinical Haematology (2nd Ed). Oxford: Oxford
University Press, 2004.
11. King M. A medical laboratory for developing countries. London: Oxford University Press, 1973.
12. Kawthalkar SM. Essentials of Haematology. New Delhi: Jaypee Brothers Medical Publishers (P) Ltd, 2006.
13. Gaw A, Murphy MJ, Cowan RA, O’Reilly DSJ, Stewart MJ, Shepherd J. Clinical biochemistry. An illustrated colour test.
3rd Ed. Edinburgh: Churchill Livingstone, 2004.
14. Hoffbrand AV, Pettit JE, Moss PAH. Essential Haematology (4th Ed). Oxford. Blackwell Science Ltd, 2001.
15. Chatterjee K, Sen A. Step by step blood transfusion services. A practical manual on the technical and clinical aspects.
New Delhi: Jaypee Brothers and Medical Publishers (P) Ltd, 2006.

tahir99 - UnitedVRG
vip.persianss.ir
Index

A Amylase 97, 135 Basal


Abnormal Anaphylactic reaction 355 acid output 123, 125
coagulation profile 66 Anemia of chronic disease 248 body temperature 156
crystals 28 Angina pectoris 76 Basic laboratory studies in anemia 260
red cell arrangement 206 Animal inoculation studies 242 Basophilic stippling 205
ABO Antibodies of ABO system 331 Basophils 174, 203, 207
grouping 336 Antibody screening and identification Bence Jones proteinuria 11
system 329 353 Benedict’s qualitative
Abortion 148 Antigens of solution 13
Acetest tablet ABO system 330 test 13
method 15 Rh system 332 Benedict’s test 14
test 15 Antiglobulin test 270 Bentiromide test 134
Acid Antiphospholipid syndrome 312 Bernard Soulier syndrome 293
Antisperm antibodies 164 Bile
load test 37
Antithyroid antibodies 143 pigment 16
phosphatase 166
Anuria 5 salts 17
Acquired
Aplastic anemia 249 Biliary peritonitis 67
disorders of coagulation 296
Apolipoproteins 71 Bilirubin 16, 97
inhibitors of coagulation 296
Appearance of sputum 99 crystals 28
Activated
Approach to diagnosis of Bioassays 148
partial thromboplastin time 303
anemia 259 Biochemical
protein C resistance 311
bleeding disorders 297 analysis of semen 165
Acute
Apt test 118 cardiac marker 74
coronary syndrome 74
Ascaris lumbricoides 112 studies 77
hemolytic transfusion reaction 354
Ascorbic acid 13 Biosynthesis of thyroid hormones 137
hepatitis 66
Asexual cycle 229 Blast cells 208
leukemias 273
Assessment of severity of Bleeding
myeloid leukemia 9
inflammatory disorders 218 diathesis 66, 82
phase reactants 218
Atypical lymphocytes 208 disorders 291
Advantages of automated hematology
Autohemolysis test 270 Blood 18
analyzer 319
Automated ammonia 59
Adverse effects of transfusion 354
hematology analyzer 319 biochemistry 33
Albumin 96 components 359, 361
method 301
Albuminuria 35 group
Automation in hematology 319
Alcoholic liver disease 66 A 330
Alicylates 13 B AB 330
Alimentary glycosuria 12 B cell B 330
Alkaline picrate reaction 34 development 175 O 330
Allergic reaction 356 ontogeny 175 systems 329, 335
Ammonium Bacteria 24, 350 mixed CSF 83
chloride loading test 37 Bacterial smear 200
magnesium phosphate 27 contamination of donor unit 355 transfusion 343
urate crystals 27 culture 81 urea nitrogen 33
Amorphous urates 27 proliferation 4 vessel wall 288

tahir99 - UnitedVRG
vip.persianss.ir
368 Essentials of Clinical Pathology

Boiling test 9 cell count in CSF 84 Clean-catch specimen 4


Bone marrow CK 78 Clearance of radiolabeled agents 32
aspiration 222, 224, 241 MCV 213 Clearance tests 32
smears 225 serum creatinine level 34 Clinitest tablet method 14
examination 266 total T4 142 Clot formation 84
iron stain 246 urobilinogen in urine 17 Clotting time 302
trephine biopsy 223, 224, 227 ketonuria 14 Coagulation system 289
Boric acid 5 low MCV 213 Coccidia 111
Breath malabsorption 118, 127 Collection methods 4
CO2 tests 131 male infertility 151 Collection of
hydrogen test 130, 131 pleural effusion 91 blood 179
tests 134 positive test 271 cerebrospinal fluid 80
Broad casts 26 prolongation of bleeding time 301 donor blood 344
Bromosulphthalein excretion test 63 proteinuria 8 pleural fluid 91
Bromothymol blue 7 Cell 22 sample 95, 123
Brugia malayi 239 count 93, 98 semen for investigation of infertility
Bulbourethral glands 159 ontogeny 175 159
Butter fat test 129 Cellular specimen for parasites 105
casts 27 sputum 99
C elements 3 urine 3
Cabot’s rings 206 Cephalic phase 121 Colorimetric methods 183
Calcium Cephalosporins 13 Combined acidity 124
carbonate crystals 27 Cerebrospinal fluid 80, 85 Commercial automated culture
hydrogen phosphate 27 systems 102
Cervical mucus penetration test 166
oxalate crystals 27 Compatibility test 352
Charcot joints 39
Capillary tube method 238 Complete blood count including blood
Cholestasis 63
Cardiac tamponade 224 smear 298
Cholesterol 69
Cardiorespiratory compromise 82 Complications associated with massive
crystals 28
Casts 25 transfusion 357
Christmas disease 294
Catabolism of steroid hormones 52 Complications of
Chromosomal analysis 152
Catheter specimen 4 bone marrow aspiration 224
Chronic
Causes of lumbar puncture 82
leukemias 280
Composition of normal
ascites 95 liver disease 66
cerebrospinal fluid in adult 80
decreased lymphocytic leukemia 283
urine 3
CSF pressure 83 myeloid leukemia 281
Computer-assisted semen analysis 166
serum creatinine level 34 renal disease 30
Concepts of universal donor and
total T4 143 Chylomicrons 69
recipient 332
erroneous results 323 Chylothorax 94
Congo red test 126
false Classic nitroprusside reaction 15
Conjugated
negative test 20, 118, 352 Classical hemophilia 294
bilirubin 56, 58
positive test 19, 118, 352 Classification of hyperbilirubinemia 56
female infertility 154 acute leukemias 273 Contraindications to
glycosuria 12 anemias 244 gastric analysis 123
hematuria 18 diabetes mellitus 39 lumbar puncture 82
hemoglobinuria 20 intestinal parasites of humans 107 Coombs’ test 270
increased lipoprotein disorders 72 Copper reduction
ALP 61 liver function tests 54 methods 13
BUN 34 renal function tests 30 tablet test 14

tahir99 - UnitedVRG
vip.persianss.ir
Index 369

Correction for Detection of Double oxalate 182


abnormal solute concentration 7 antigen in stool samples 111 Drugs 12
dilution 7 filarial Dubin-Johnson syndrome 56
temperature 7 antigen 239 Duffy antigen 231
Correction of TLC for nucleated red DNA 240 D-xylose absorption test 130
cells 194 infection 218
Counting cells 194 microalbuminuria 11 E
Creatine kinase 78 nucleic acid sequences of malaria Ectopic pregnancy 147
Creatinine clearance 32 parasites 236 Eggs of schistosoma haematobium 25
Crigler-Najjar syndrome 55 parasites 104 Ehrlich’s aldehyde test 18
Criteria for selection of blood donors postoperative infection 219 Electrical
342 Determination of blood group conductivity 320
Crohn’s disease 211 substances 166 impedance 319
Cryptococcus neoformans 87 Determining cause of anemia 260 Emergency cross-match 353
Cryptosporidium parvum 111 Diabetes mellitus 4, 12, 39-41, 44 Endocrine
Crystals 27 Diacetyl monoxime urea method 34 component 133
present in Diagnosis of diseases 12
acid urine 27 malaria and other parasites in blood Endocytosis of colloid droplets 138
alkaline urine 27 229 Endogenous pathway 71
CSF renal disease 30 Endometrial biopsy 155
culture 88 Diagnostic thoracentesis 91 Endoscopic biopsy of ulcer in intestine
protein electrophoresis 88 Diazo method 58 110
Culture 102 Differential leukocyte count 85, 208 Entamoeba histolytica 108
Cushing’s syndrome 12 Diffusion of iodide 137 Enterobius vermicularis 114
Cyanmethemoglobin method 185 Dilution of blood 193 Enzymatic methods 35
Cyclospora cayetanensis 111 Direct Enzyme-linked immunosorbent assay
Cystatin C clearance 32 antiglobulin test 270 348
Cysteine crystals 28 bilirubin 58 Eosinophil 174, 203, 207
Cytogenetic analysis 280 fluorescent antibody assay 111 Eosinophilia 210
Cytological examination of sputum spectroscopic estimation 58 Epididymis 159
103 tests 133 Errors in
Cytoplasmic vacuoles 208 wet mount of CSF 87 blood collection 194
Directed donor 342 filling of chamber 194
D Disadvantages of automated pipetting 194
D-dimer test 94 hematology analyzer 319 Erythrocyte sedimentation rate 215,
Decrease in glucose 4 Disintegration of cellular elements 4 219
Decreased utilization of carbohydrates Disorders of Erythrocytic schizogony 230
14 blood vessels 292 Erythropoiesis 171
Deficiency of lipids 69 Escherichia coli 6, 7
antithrombin III 312 platelet 292, 293 Esophageal disease 94
protein C and S 312 thyroid 139 Esophagogastroduodenoscopy 126
Delayed hemolytic transfusion reaction Disseminated intravascular Estimation of
355 coagulation (DIC) 296 blood glucose 45
Demonstration of Distal tubular function 36 creatinine clearance from serum
eggs of A. lumbricoides 112 DNA diagnosis 242 creatinine 33
hookworm eggs 113 Döhle inclusion bodies 207 fecal
trophozoites 110 Donation interval 342 enzymes 134
Dental treatment 343 Donor reactions 345 fat 129, 135

tahir99 - UnitedVRG
vip.persianss.ir
370 Essentials of Clinical Pathology

glucose in CSF 86 Fecal Gilbert’s syndrome 55


hemoglobin 183, 268, 321, 344 fat 129 Glanzmann’s thrombasthenia 294
progesterone in mid-luteal phase osmotic gap 119 Glitter cells 24
157 pH 119 Glomerular proteinuria 8
protein 11 Female infertility 154 Glomerulonephritis nephrotic
red blood cell count 321 Fern test 156 syndrome 3
red cell distribution width 321 Ferric chloride test 15, 16 Glucose 12
Ethylene diamine tetra-acetic acid Fibrinolytic system 290 6-phosphate dehydrogenase
(EDTA) 181 Flagging 323 deficiency 231, 255
Evaluation of Floatation techniques 107 tolerance test 12
chronic diarrhea 104 Florence test 166 Glycated hemoglobin 47
clinical features in anemia 260 Flow cytometric analysis 272 Glycosuria 6, 35, 43, 48
dysentery 104 Fluorescence 320, 325 without hyperglycemia 12
malabsorption syndromes 127 microscopy 101, 236 Gmelin’s test 16
Examination of Foam test 16 Gram’s
ascitic fluid 96 Follicle stimulating hormone 150, 151 stained smear 25
blood smear 203 Follicular cell and proteolysis 138 smear 87
bone marrow 220 Formalin 5 Granular casts 26
cerebrospinal fluid 80 Formation of crystals 4 Graves’ disease 144
feces 104 Fouchet’s test 17 Gross appearance of cerebrospinal
marrow specimens 225 Fractional fluid 83
pleural fluid 93 excretion of sodium 35
seminal fluid 160 test meal 126 H
splenic aspirate 241 Haemophilus influenzae 82, 88, 100
Free
sputum 99 Ham’s acidified serum lysis test 271
acidity 124
smear 101 Hamster egg penetration assay 166
thyroxine 143
urine 3 Hay’s surface tension test 17
Fresh frozen plasma (FFP) 362
Excretion of metabolic waste products HDL-cholesterol 74
Froin’s syndrome 84
30 Heat and acetic acid test 9
Frozen red cells 361
Excretory function 52 Heinz bodies 199, 269
Functions of
Exocrine component 131 Helminths 112
cerebrospinal fluid 80
Exoerythrocytic schizogony 230 Hematopoiesis 52, 169
liver 52
Exogenous pathway 71
Further testing 318 Hemiglobincyanide method 185
Extrahepatic biliary obstruction 66
Hemocytometer 192
Exudates 93 G Hemodynamic proteinuria 9
F Gametogony 230 Hemoglobin 20, 172
Gasometric method 183 content 204
Factor V Leiden 311
Gastric D 254
Factors affecting
analysis 121 electrophoresis 267
erythrocyte sedimentation rate 215
phase 121 Hemolysis 17
renal function 30
Gastrin 132 Hemolytic
False reactions in ABO grouping 338
Fasting Gaucher’s disease 221 anemia 261
blood glucose 45 General metabolic functions 52 disease of newborn 256
glucose 3 Generalized aminoaciduria 35 uremic syndrome 293
plasma glucose 47 Gestational Hemophilia
Fatty casts 26 diabetes mellitus 41 A 294
Febrile non-hemolytic transfusion trophoblastic disease 148 B 294
reaction 355 Giardia intestinalis 110 Hemorrhage 18, 224

tahir99 - UnitedVRG
vip.persianss.ir
Index 371

Hemorrhagic Hypothyroidism 140 semen analysis 159


disease of newborn 296 Hysterosalpingo-contrast sonography testing for thrombophilia 313
fluid 93, 96 157 urinalysis 3
Hemosiderin 20 Hysterosalpingography (HSG) 157 Indirect
Heparin 182 antiglobulin test 271, 353
induced thrombocytopenia 312 I bilirubin 58
Hepatic Identification of tests 134
injury 59 adult worms 112 urinary tract infection 21
jaundice 55 cause of dyslipidemia 74 Induction of sputum 99
schizogony 230 larvae of S. stercoralis 114 Infants 4
Hepatitis lipid disorder 73 Infection in spinal canal 82
A virus 356 malaria parasites 232 Infectious disease 157, 343
B virus 348, 356 rotavirus 105 Infertility 150
C virus 349, 356 trophozoites and cysts on stool 108 Infiltrative diseases 63
Hepatocellular injury 63 Iliac spines 222 Inherited
Hereditary Illness 343
disorders of coagulation 294
disorders of hemoglobin 250 Immediate spin cross-match 352
thrombophilia 311
spherocytosis 254 Immune
Insulin
Herniation of brain 82 hemolytic anemias 255
hypoglycemia test 125
High thrombocytopenic purpura 292
resistance syndrome 42
density lipoprotein 71 Immunochemical tests 118
Intermediate density lipoprotein (IDL)
fever 9 Immunoelectrophoresis 285
70
performance liquid Immunofixation electrophoresis 286
Interpretation of
chromatography 268 Immunological assays 148
liver function tests 63
Histoplasma capsulatum 212, 226 Immunophenotyping 278
screening tests 306
Histoplasmosis 221 Importance of HLA antigens 177
Intrahepatic cholestasis 56
Hodgkin’s disease 211 Increase in pH 4
Intraperitoneal hemorrhage 67
Hollander’s test 125 Increased
Intravascular hemolysis 9, 264
Hookworms 113 destruction in peripheral blood 210
Inulin clearance 32
Hormonal studies 152 sequestration in spleen 210
Investigation of
Howell-Jolly bodies 199, 205 soluble transferrin receptor 246
male infertility 152
Human testosterone 157
pyrexia of unknown origin 221
albumin solutions 362 Indications and limitations of liver
function test 53 Iron
chorionic gonadotropin 146
Indications for deficiency anemia 245
cycle 229
abdominal paracentesis 95 overload 356
immunodeficiency virus 343, 349,
bone marrow staining of bone marrow aspiration
356
aspiration 221 smears 226
leukocyte antigens 175
biopsy 221 studies 265
Hydatid cyst of liver 67
examination 221 Irradiated red cells 361
Hydrochloric acid 5, 122
gastric analysis 122 Isomorphic red cells 23
Hyperglycemia 43
Hyperhomocysteinemia 312 hemoglobin 183 Isoniazid 13
Hyperosmolar hyperglycemic state lumbar puncture 81 Isospora belli 111
(HHS) 44 measurement of erythrocyte Ivy’s method 301
Hypersegmented neutrophils 208 sedimentation 216
Hyperthyroidism 139 renal J
Hypertonic urine 22 biopsy 37 Jaffe’s reaction 34
Hypo-osmotic swelling of flagella 166 function tests 30 Jaundice 54

tahir99 - UnitedVRG
vip.persianss.ir
372 Essentials of Clinical Pathology

K Life cycle of Mediastinitis 224


Kala azar 221 Leishmania donovani 240 Megaloblastic anemia 246
Karyopyknotic index (KI) 156 malaria parasites 229 Membrane filtration 238
Kawamoto technique 236 Light Metabolic
Ketones 14 absorption 320 actions of insulin 39
Ketosis 43 scatter 325 alterations in diabetes mellitus 43
Kidney function 30 Lipoproteins 69 syndrome 42
Klebsiella pneumoniae 100 Liquid biopsy of urinary tract 21 Metabolism of thyroid hormones 138
Litmus paper test 7 Method of gastric analysis 123
L Liver Methods for estimation of
Laboratory diagnosis of biopsy 64 BUN 34
acute leukemias 275 disease 296 erythrocyte sedimentation 216
visceral leishmaniasis 241 function tests 52 hemoglobin 183
cerebrospinal fluid 82 transplantation 66 serum creatinine 34
Laboratory testing in thrombophilia Loeffler’s syndrome 210 Methods for HLA antigen typing 177
313 Loop of Henle 8 Micro method 189
Laboratory tests for Loss of ketone bodies 4 Microalbuminuria 11, 35, 49
diagnosis of diabetes mellitus 44 Low-density lipoprotein (LDL) 71 Microangiopathic hemolytic anemia
human chorionic gonadotropin 148 Lugol iodine test 16 258
lipoprotein disorders 73 Lundh meal 133 Microangiopathy 44
screening of diabetes mellitus 47 Lung diseases 210 Microcytic hypochromic anemia 221,
Laboratory tests in Lupus pleuritis 94 261
anemia 244 Luteinizing hormone 150 Microfilariae 25
bleeding disorders 288 Lymphatic filariasis 237
Microhematocrit tube 238
hematological malignancies 273 Lymphocytes 175, 207
Microplate method 338
management of acute metabolic Lympocytotoxicity test 177
Microscopic examination of
complication of diabetes mellitus 50 Lysed
blood for demonstration 238
porphyrias 314 capillary blood method 238
urinary sediment 19
thrombophilia 311 venous blood method 238
Microsporidia 111
Laboratory tests to assess
M Microvascular disease 44
glycemic control 47
Midstream specimen 4
long-term risks 49 Macro method 188
Miliary tuberculosis 221
Lactate dehydrogenase 97 Macroangiopathy 44
Macrocytic anemias 261 Mixed lymphocyte
Lactation 343
Macrophages 52 culture 177
Lactose tolerance test 130
Macrovascular disease 44 reaction 177
Laparoscopic liver biopsy 67
Malabsorption syndromes 127 Modes of transmission 231
Laparoscopy and dye hydrotubation
Malaria 229, 343 Molecular genetic
test 157
Male infertility 150 analysis 280
Latex agglutination tests 87, 88
Manual method 192, 299 techniques 177
LDL-cholesterol 74
Mastalgia 155 Molecular methods 102
Leishman stain 202
Maximum acid output (MAO) 123 Monocytes 174, 203, 207
Leishmania donovani 211, 226
Mean cell Moraxella catarrhalis 100
Leucine crystals 29
hemoglobin 213 Morphological classification of anemia
Leucocyte esterase test 21
Leukemoid reaction 209 concentration 214 213
Leukocyte-poor red cells 361 volume 213 Morphology 275

tahir99 - UnitedVRG
vip.persianss.ir
Index 373

Morphology of Oral Phosphates 27


abnormal leukocytes 207 contraceptive therapy and Physiologic jaundice of newborn 56
microfilariae on Romanowsky pregnancy 312 Physiology of hemostasis 288
stained 238 glucose tolerance test 45 Plasma
normal leukocytes 207 Organ transplantation 177 cell dyscrasias 9, 283
Mosquito cycle 230 Organisms 24, 211 components 362
Mycobacterium tuberculosis 87, 101 Orthostatic proteinuria 3 derivatives 362
Myeloperoxidase (MPO) 276 Osmotic fragility test 269 proteins 289
Myocardial infarction 219 Ounting chamber with cover glass 192 Plasmodium species 351
Oval fat bodies 24 Platelet 203, 211, 288, 361
Myoglobin 20, 78
Overflow proteinuria 9 concentrate 361
N Oxidation of count 299, 322
Naegleria fowleri 87 bilirubin to biliverdin 4 function analyzer 302
urobilinogen to urobilin 4 glycoprotein analysis 310
Nalidixic acid 13
Oxyhemoglobin method 186 pheresis 362
NBT-PABA test 134
Neimann-Pick disease 221 Pleural
P
Neisseria meningitidis 82 biopsy 94
P. falciparum 231
Neonatal screening for hypothyroidism effusion 91
P. ovale 231
144 fluid 91
P. vivax 231
Nephropathy 39 Polychromatic cells 203
Packed
Nephrotic syndrome 6 Polymerase chain reaction 87, 88, 177
cell volume 188
Polymorphonuclear neutrophil 207
Neurogenic phase 121 red cells 361
Polyuria 5
Neutropenia 210 Pancreas 131
Postcoital test 165
Neutrophilia 209 Pancreatic
Posthepatic jaundice 56
Neutrophils 174, 203 disease 12, 94
Postprandial blood glucose 45
Nitrite test 21 function tests 131
Post-puncture headache 82
Nitroprusside test 16 Pancreolauryl test 134
Post-renal proteinuria 9
Non-cellular casts 26 Pappenheimer bodies 199, 205
Post-transfusion purpura 293
Non-endocrine diseases 12 Para-aminosalicylic acid 13
Postural proteinuria 9
Non-specific esterase (NSE) 276 Parasite culture 242
Prediabetes 42
Normal Parasites 203, 351
Paroxysmal nocturnal hemoglobinuria Pre-erythrocytic schizogony 230
absorption of carbohydrates 127
258 Pregnancy 343
bilirubin metabolism 52 test 3, 146
bone marrow 220 Peak acid output 123, 125
Pelger-Huet cells 208 Prehepatic jaundice 55
crystals 27 Preparation of
Penicillins 13
gastric anatomy and physiology 121 blood smear 200
Percutaneous
pregnancy 146 slides 106
blind liver biopsy 66
Normocytic normochromic anemia 261 Preservation of urine sample 4
guided liver biopsy 67
Numerical abnormalities of leukocytes Primary
plugged liver biopsy 67
209 biliary cirrhosis 56, 66
Periodic acid Schiff (PAS) 278
Peripheral sclerosing cholangitis 56, 66
O blood smear 260 Primed lymphocyte typing 177
Obstructive jaundice 18 neuropathy 39 Processing of
OGTT in gestational diabetes mellitus Peritoneal fluid 95 donor blood 345
46 pH marrow specimens 224
Oligoclonal bands 89 indicator paper 7 Production of vit. D3 30
Oliguria 5 meter 7 Professional donors 342

tahir99 - UnitedVRG
vip.persianss.ir
374 Essentials of Clinical Pathology

Prostate 159 Reduction of intestinal bacterial flora Self-monitoring of blood glucose


Proteins 8, 97 18 (SMBG) 48
Proteinuria 6 Refractometer method 7, 58 Semen 159
Prothrombin Regular cycles 155 analysis 152, 159
complex concentrate 363 Regulation of blood pressure 30 Seminal
time 59, 302 Renal fluid 159
Protozoa 107 biopsy 37 vesicles 159
Pseudochylous effusion 93 failure 3 Sensitivity of test 13
Pseudomonas aeruginosa 100 function tests 30 Sequence of filling of tubes 182
Pus cells 23 transplantation 30 Serologic
Pyelonephritis 3 tubular epithelial cell 24, 27 methods 237
Pyloric part 121 Replacement blood donor 342 tests 110, 87, 88
Pyrexia 66 Response to oral iron therapy 246 Serological testing 177
Restriction fragment length Serum
Q polymorphism 177 albumin 59
Qualitative tests 147 Reticulocyte count 196261, 323 alkaline phosphatase 61
Quantitative Retinopathy 39 aminotransferases 60
buffy coat 236 Reverse passive hemagglutination bilirubin 56
estimation of assay 348 creatinine 34, 35
fecal fat 118 Reye’s syndrome 59 ferritin 245
proteins 10 Rh iron 245
antibodies 335 lipase 135
tests 147
D grouping 338 protein electrophoresis 59, 284
Queckenstedt’s test 81
system 332 triglycerides 73
R Rheumatoid effusion 94 Sexual cycle 230
Radioactive iodine uptake test 144 Role of Sickle cell
Random genetic factors in malaria 231 disorders 253
laboratory tests in diabetes mellitus slide test 266
blood glucose 45
44 Sideroblastic anemia 249
donor platelets 361
Rothera’s Significance of
Reaction and pH 7
nitroprusside method 15 erythrocyte sedimentation rate 215
Reagent strip
test 15 microalbuminuria 11
method 7, 14, 18
Rouleaux formation 338 Sims-Huhner test 165
test 7, 10, 15, 16
Round cells 163 Single
Red blood cells 22, 171
Roundworm 112 donor platelets 362
Red cell 203
radial immunodiffusion 286
casts 27 S Sites for bone marrow aspiration 222
components 361 Sahli’s acid hematin method 184 Skeletal muscle trauma 9
distribution width 214 Scattergram 323 Skin
enzymes 172 Schizogony 229 piercing 343
inclusions 205, 268 Screening tests for puncture 179
indices 213, 261 hemostasis 298 test 242
membrane 172 infections transmissible by Slide test 336
suspension 361 transfusion 347 Small lymphocytes 203
Red cells in additive solution 361 Secondary lipoprotein diseases 72 Solubility test for hemoglobin S 266
Red cells with abnormal Secretin-cerulin test 133 Sources of error in manual blood cell
shape 205 Sedimentation count 194
size 204 rate 215 Specific gravity 6
staining 204 techniques 107 method 186

tahir99 - UnitedVRG
vip.persianss.ir
Index 375

Specific tests for hemostasis 307 T pancreatic function 133


Sperm T cell paroxysmal nocturnal
count 162 development 175 hemoglobinuria 271
function tests 165 ontogeny 175 porphyrins in erythrocytes and
morphology 162 Taenia plasma 318
motility 161 saginata 116 Thalassemias 250
viability 161 solium 115 Therapeutic thoracentesis 92
vitality 161 Tallqvist hemoglobin chart 184 Thick
Spermatozoa 24, 159 Tamm-Horsfall protein 8, 25 blood smear 238
Sperms 159 Telescoped urinary sediment 24 exudative fluid 93
Spinnbarkeit test 156 Tense ascites 67 viscous CSF 84
Spinous processes of lumbar vertebra Terminology in flow cytometry 324 Thoracentesis 91
222 Test for Thoracoscopy 95
Sporogony 230 fructose 165 Thrombin time (TT) 305
Spot check of gastric pH 126 malabsorption of fat 118 Thrombopoiesis 178
Squamous epithelial cells 24 occult blood in stools 117 Thrombotic thrombocytopenic
Stages of pancreatic arylesterase 134 purpura 293
erythrocyte sedimentation rate 215 porphobilinogen in urine 318 Thymol 5
iron deficiency 245 reducing sugars 119 Thyroid
Staining of blood smear 202 total porphyrins in function tests 137, 142
Staphylococcus aureus 100 feces 318 hormones 137
Statistical error 194 urine 318 scintiscanning 144
Stellar phosphate 27 urobilinogen in feces 119 stimulating hormone 142
Sterile’ pyuria 25 Testes 159 Thyrotropin releasing hormone
Sternum 222 Testicular biopsy 152 stimulation test 143
Straw-colored transudative fluid 93 Testing for ketone bodies 51 Tibia 222
Streptococcus Tests for detection of Time of collection 3
pneumoniae 100 anti-leishmania antibodies 241 Titration 124
pyogenes 100 bilirubin in urine 16 Toluene 5
Streptomycin 13 blood in urine 19 Total
Strongyloides stercoralis 113 glucose in urine 13 acidity 124
Subarachnoidal epidermal cyst 82 hemoglobinuria 20 cholesterol 73
Subdural hematoma 82 ketones in urine 15 iron binding capacity 245
Sucrose hemolysis test 271 occult blood in feces 117 leukocyte count 84, 192
Sulfonamide crystals 29 urobilinogen in urine 18 serum proteins 58
Sulphosalicylic acid test 10 Tests for thyroxine 142
Suspected evaluate Toxic granules 207
acute leukemia 221 glomerular function 30 Transferrin saturation 245
chronic lymphoid leukemias 221 tubular function 35 Transfusion associated lung injury 355
infections 221 gastric analysis 125 Transitional epithelial cells 24
myelodysplastic syndrome 221 glucose-6-phosphate Transvaginal ultrasonography 157
myeloproliferative disorders 221 dehydrogenase deficiency 270 Transvenous liver biopsy 67
plasma cell dyscrasia 221 hemoglobin S 266 Treponema pallidum 350, 357
storage disorder 221 malabsorption and pancreatic Trichomonas vaginalis 25
Synthesis of erythropoietin 30 function 127 Trichuris trichiura 113
Synthetic function 52 ovulation 155 Triglycerides 69

tahir99 - UnitedVRG
vip.persianss.ir
376 Essentials of Clinical Pathology

Triple phosphates 27 Urinometer method 6 Waxy cast 26


Trisodium citrate 182 Urobilinogen 17 WBC cytogram 323
Troponins 78 Uses of Wedge method 200
True chylous effusion 93 C-reactive protein 218 Westergren method 216, 219
Trypanosomes 87 PCV 188
White
Tube method 337 red cell indices 213
blood cells 23, 174
Tubeless gastric analysis 126
V cell 203, 206
Tuberculous pleural effusion 94
Vagina 166 casts 27
Tubes for collection of blood 182
Tubular proteinuria 9, 35 Vaginal cytology 156 count 96
Turk solution 84 Vas deferens 159 WHO hemoglobin color scale 185
Types of Vasectomy 166 Wilson’s disease 66
blood donors 341 Venous blood collection 179 Wintrobe
liver biopsy 65 Very low-density lipoproteins (VLDL) method 188, 218
Tyrosine crystals 29 70 mixture 182
Viral hepatitis 343 Wuchereria bancrofti 238
U Viruses 348
Ultrasonography (USG) 156 Visceral leishmaniasis 240 X
Unconjugated bilirubin 58 Viscosity 161 Xanthochromia 84
Unexplained cytopenia 221 Vitamin
Unstable angina 76 B12 and folate 265 Y
Urea clearance 33 K deficiency 296
Yeast cells 25
Urease-berthelot reaction 34 Volume of donation 343
Yersinia pestis 100
Uric acid crystals 27 von Willebrand disease (vWD) 294
Urinary Z
albumin excretion 49 W
Zeta sedimentation ratio (ZSR) 218
concentration of sodium 35 Washed red cells 361
Water Ziehl-Neelsen
Urine
bilirubin and urobilinogen 58 deprivation test 36 smear 87
osmolality 36 loading antidiuretic hormone technique 101
specific gravity 36 suppression test 36 Zollinger-Ellison syndrome 122

tahir99 - UnitedVRG
vip.persianss.ir

You might also like