You are on page 1of 83

EMILIO AGUINALDO COLLEGE, MANILA

1113-1117 San Marcelino St. corner Gonzales St., Ermita, Manila


School of Medical Technology

Hematology synthesis
Submitted by :
Ibrahim , Riman Mustafa
I. HEMATOPOIESIS
• Stem cells are defined as cells that are • HSCs differentiate into multipotent
capable of self-renewal and progenitors (MPPs), which do not have the
differentiation into multi-lineage cells capacity to self-renew but can generate
Types of human steam cell lineage-committed progenitors, including

1.Totipotential stem cells A) common lymphoid progenitors (CLPs)


• Present in the first few hour after an
ovum is fertilized - that differentiate into T-lymphocytes, B-
• Most versatile type of stem cell lymphocytes, and natural killer cells.
• Can develop into any human cell type ,
including development from embryo into B) common myeloid progenitors (CMPs)
fetus
- CMPs give rise to granulocyte/macrophage
2. Pluripotential stem cells progenitors (GMPs) - mature granulocytes,
• Present several days after fertilization monocytes and macrophage
• Can develop into any cell type except
they cant develop into fetus - megakaryocyte/erythrocyte progenitors
(MEPs) - differentiate into platelets and
3.Multipotential stem cells erythrocytes
• Developed from pluripotential stem cells
• Can be found in adults but they are HEMATOPOIETIC GROWTH FACTORS
limited to specific type of cells to form • Differentiation and maturation of
tissues hematopoietic cells are influenced
by soluble factors, including
• .Embryonic stem cells are pluripotent and Hematopoietic growth factors and
have the ability to generate all tissues in cytokines
the body • They regulate the proliferation, survival,
and differentiation of hematopoietic
• Hematopoietic stem cells (HSCs) are precursor cells and facilitate the function
characterized by their ability to self of mature blood cells.
renew and differentiate into committed • They are produced by different types of
hematopoietic progenitors. hematopoietic or non-hematopoietic cells
• HSCs give rise to all mature blood cells of and usually affect more than one lineage
the hematopoietic system
• Another class of factors, called
chemokines, is important in regulating
hematopoietic cell trafficking and homing
• Erythropoietin – stimulates proliferation , • At the sixth week, hematopoiesis begins
growth and differentiation of erythroid in the liver, and this becomes the major
precursor , produced by the kidneys. hematopoietic organ of early and
• Thrombopoietin – growth factor that midfetal life.
primary regulator of platelet production ,
produced by the kidney. • Definitive erythroblasts, which become
• Granulocyte-Monocyte colony nonnucleated red cells, are formed
stimulating factor (CSF-MG)- a myeloid extravascularly in the liver, and
growth factor that stimulates erythroid , granulopoiesis and megakaryocytes are
granulocyte , monocyte , megakaryocyte , present to a lesser degree
eosinophil progenitor .
• Granulocyte- colony stimulating factor • In the middle part of fetal life, the spleen
(CSF-G)- stimulate granulocyte , lymph nodes thymus and kidney have a
production and functional activation minor role in hematopoiesis, but the liver
continues to dominate
II. HEMATOPOIETIC TISSUES Note : major hemoglobin in this phase is HbF
-The 3 phases and sIte of Hematopoiesis

A) EMBRYONIC hematopoiesis (mesoblastic


period )- 19th- 20th day of gestation upto 8th- C)POSTNATAL HEMATOPOIESIS (medullary/
12thweek myeloid phase)
• Shortly after birth, hematopoiesis in the
-Beginning in the first month of prenatal life, liver ceases, and the bone marrow is the
primitive hematopoiesis starts outside the only site for the production of
embryo in the mesenchyme of the yolk sac as erythrocytes, granulocytes, and platelets.
blood islands

✓ - The first blood cell produced are • Hematopoietic stem cells and committed
RBC but its called primitive progenitor cells are maintained in the
erythroblasts, which are megaloblastic , marrow
only during this phase they take place
intravascularly. • Lymphocytes (Bcells ) continue to be
produced in the marrow, as well as in the
Note : major hemoglobin in this phase are – secondary lymphoid organs, &T
Gower1, Gower 2 and Portland lymphocytes are produced in the thymus
and also in the secondary lymphoid
B) FETAL HEMATOPOIESIS (liver phase) - organs
• Bone marrow contains two types of stem
• Definitive hematopoiesis in mammals cells: hemopoietic (which can produce
blood cells) and stromal (which can
takes place in the aorta-gonad
produce fat, cartilage and bone
mesonephros (AGM) region of the
embryo, from which hematopoietic cells
migrate to the placenta, liver, and
spleen.
• There are two types of bone marrow: red
marrow (also known as myeloid tissue) • Begins to accumulate component for
and yellow marrow. hemoglobin production
• At birth, the total marrow space is • The pronormoblast undergoes mitosis
occupied by active hematopoietic (red) and forms two basophilic normoblasts
marrow (capable of cell division )
• Red marrow is found mainly in the flat • Present only in bone marrow
bones such as hip bone, breast bone, skull,
ribs, vertebrae , thoracic , pelvis and
shoulder blades, and in the cancellous 2) Basophilic Normoblast
("spongy") plus the proximal parts of the • Nucleoli are present but are not often
long bones of the upper and lower limbs
visible
• The nuclear/ cytoplasmic (N/C) ratio is
• 5th - 7th years of age - only part of that
moderate (6:1)
space is needed for hematopoiesis; the
• cytoplasm is deeply basophilic, owing to
remaining space is occupied by fat cells
the abundance of RNA (darker than
(yellow marrow) which are inactive cause
pronormoblast)
mainly they are made of Adipocytes
• slightly coarser chromatin that stains
intensely; the chromatin may be partially
• yellow marrow that can be replaced by
clumped, and the pattern may suggest a
hematopoietic cells if continuous,
wheel with broad spokes.
intensive stimulation exists.
• Capable of cell division & present only in
BM
• Shifting of Red marrow to Yellow marrow
• Detectable Hb synthesis occurs but
as body growth progress is called
completely masked by large number of
Retrogression processes
RNA and ribosomes. (1st cell capbale of
Hb synthesis )
Note : this process takes place
extravascularly and there will be shift of the
3) Polychromatophilic normoblast
concentration of HbF to HbA and HbA2
• slightly smaller than the basophilic
normoblast
III. ERYTHROPOIESIS
1) Pronormoblast • nucleus occupies about half of the area
of the cell, stains intensely
• earliest recognizable erythroid precursor
• nucleus occupies about half of the area
• it is the largest of the erythroid precursor
of the cell, stains intensely (4:1)
high nucleus to cytoplasm ratio
• Blue greyish to pink grey cytoplasm
• nucleus has a fine, uniform chromatin
pattern that is somewhat more distinct • After mitosis ,Last cell capable of
• Blue cytoplasm, intensely stained , due to mitosis, the nucleus becomes small and
dense (pyknotic)
high concentration of ribosome
4) Orthochromatic normoblast • In the marrow, developing erythroid
• Completely condensed nucleus (non cells are usually in contact with
functional ) macrophages in what are termed
• The cytoplasm contains more abundant erythroblastic islands
hemoglobin and fewer polyribosome
• Cytoplasm appears pink b/c its fully • If sufficiently severe hypoxia is present,
hemoglobinized this marrow pool of reticulocytes can be
• Not capable of cell division cause released. This approximately doubles the
chromatin is condensed number of circulating reticulocytes .
• Nucles is ejected • They are the earliest blood cells in the
• Howell-jolly bodies (DNA remnants lineage of erythropoiesis series that are
maybe seen) seen normal in blood circulation
• Shift cells / stress cells – reticulocyte that
5) Polychromatophilic erythroblast (reticulocyte) are found or seen in case of anemia .
• the reticulocyte is polychromatophilic as Premature release of reticulocyte
a result of the retention of RNA.
• It only stains with supravital stain , 6) Erythrocytes
Romanowsky stain, • Biconcave disc shape ( allows optimal gas
• Has no nucleus exchange) non nucleated
• Completes the reproduction oh Hb • Appear salmon pink with central pallor
• Stays 1 day or longer in BM and 1 day in • Membrane is flexible and deformable
circulation that allows them to squeeze through
blood vessels
Reminder : The pronormoblast and the basophilic • circulate for about 120 days
normoblast have the highest content of RNA,
which begins to decline in the polychromatophilic Substances Needed for Erythropoiesis
normoblasts as hemoglobin increases in amount. 1. Iron: Must be in the ferrous state (Fe2+) to
Synthesis of RNA gradually decreases in each transport oxygen
stage through the orthochromatic normoblasts. 2. Amino acids: Globin-chain synthesis
When the nucleus is no longer present (in the 3. Folic acid/vitamin B12: DNA replication/cell
reticulocyte), RNA synthesis ceases, yet the RNA division
already present remains for a few days, and 4. Others: Erythropoietin, vitamin 65
protein and heme synthesis continue in the (pyridoxine), trace minerals
reticulocyte until the cell loses its RNA and
mitochondria.
Erythroid precursors or Nomenclature system

A) Pronormoblast Rubriblast Proerythroblast


B) Basophilic normoblast Prorubricyte Basophilic Erythroblast
C) Polychromatophilic normoblast Rubricyte Polychromatic erthyoblast
D) Orthochromatic normoblast Metarubricyte Orthpchromatic Erythroblast
E) Polychromatophilic erythroblast Polychromatophilic erythroblast Polychromatophilic
erythroblast
F) Erythrocyte Erythrocyte Erythrocyte

IV) SYNTHESIS OF HEMOGLOBIN

• Hemoglobin – transports oxygen Reminder : I Hb molucles holds 4 Oxygen


(oxyhemoglobin) from the lungs to the 1 heme holds – 1 oxygen (4O2 cause
tissues and carbon dioxide we have 4 heme .
(carbaminohemoglobin) from the tissue 1 oxygen holds 4 heme
to lung.
• It’s the main cytoplasmic component of • Heme synthesis occurs in mitochondria
RBC and cytoplasm RBC precursor
• Conjugated globular proteins consist of
- 4 heme group emedded in 2) Globin Synthesis
polypeptide chains
- 2 heterogenous pairs of polypeptide • Globin synthesis occurs in the cytoplasm
Chain (4 total) of the normoblast and reticulocyte.
• Composed of 2 identical pairs of unlike
1) HEME SYNTHESIS polypeptide
• Heme synthesis occurs in most cells of • 141-146 amino acids
the body, except the mature • Polypeptide chains are manufactured on
erythrocytes, but most abundantly in the Ribosomes of normoblast cytoplasm
erythroid precursors. • Globin chains are assembled from 2 pairs
• GIVES THE RBC ITS RED COLOR ( 4 chains per Hb moleucle)
• Made of Protoporphyrin IX with divalent
ferrous iron (Fe2+) 4 Globin Chain
• Protoporphyrin is normally found in 1 pairs – alpha and 1 –pair non alpha
mature erythrocytes. In lead poisoning
and in iron deficiency, levels of free Reminder :
erythrocyte protoporphyrin (FEP) are ✓ Chr 16 = alpha and zeta
increased. ✓ Chr 11 – beta , Delta , Epsilon ,Gamma
• First heme is produced then globulin
Hemoglobin Assembly

• Each globin chain binds to heme molecule and pair off . They combine to form heterodimer and
tetramer form
• Hemoglobin Ontogeny

Embryonbic Gower 1 2 epsilon + 2 zeta


Gower 2 2 aplha + 2 epsilon
Portland 2 gamma + 2 zeta
Fetal Hemoglobin F 2 alpha + 2 gamma

At birth Hb F 2 alpha + 2 gamma


Hb A
Adulthood HbA 2 alpha +2 beta
HbA2 2alpha + 2 delta
HbF 2 alpha + 2 gamma

Oxygen dissociation curve • 2,3 DPG – changes Hb molecules from


• The sigmoid-shaped oxygen dissociation relaxed oxygenated to tensed
curve of hemoglobin reflects this deoxygenated form which is stabilized by
increasing affinity for oxygen with salt bridge .
increasing partial pressure of oxygen in • Deoxygenated Hb has little affinity to
the lungs .
oxygen .
• Oxygen affinity of hemoglobin depends
• The curve is sigmoidal and shifts
on Partial Pressure of Oxygen
to the right with increasing
• PO2 and oxygen affinity relationship is
described by oxygen dissociation curve concentrations of 2,3-DPG; this
• 27mmHg is required to saturate Hb with results in decreased affinity of
50% O2 under normal condition (P50 hemoglobin for oxygen and
value ) ,Ph- 7.4 & Temp-37.5 increased delivery of oxygen to
the tissue
Shift to the left
• Increase Ph ( alkalosis decrease H+)
• Decrease 2,3 DPG IV. Hemoglobin Variants
• Decrease CO2 • Nomral Hb
• Decrease tempreature
• PO2 =<27mmHg 1) Oxyhemoglobin
✓ Hb + Oxygen , Arterial blood (bright blood
Shift to the right )
• Decrease Ph( acidosis increase H+) ✓ -is the primary Hgb for gas transport in
• Increase 2,3DPG
the body
• increase CO2
• increase temperature
2) Deoxyhemoglobin (Hb w/ oxygen )and
• Abnormal Hb w/ low affinity to O2
Carbaminohemoglobin
• PO2 =>27mmHg
✓ (Hb w/ CO2) - venous blood both forms
dark purple color to blood

• Abnormal Hemoglobin whose structure • Small amounts of carboxyhemoglobin are


has been modified by environmental , produced within the cells,but most problems
chemicals or drugs occur as a result of environmental exposures
• Blood is cherry red and measured at 540nm
1) Methemoglobin • Treatment is by hyperbaric oxygen therapy
• Hgb with oxidized iron Fe3+ (ferric state ) • Its also known as the silent killer cause its
, it is usually kept from overproduction odorless , colorless gas and may quickly
because of the methemoglobin reductase develop to hypoxia
pathway. RR – 0.2-0.8 %
• Brownish to bluish color of blood
• >30% hypoxia and cyanosis
• >50% cause death V. RED BLOOD CELL METABOLISM AND
• Increased conc. Produce a left shift PHYSIOLOGY
• Measured at 630 nm ( co-oximeter)
• Sample should be heparinized •Mature RBCs have no mitochondria, sorely on
• Treated by ascorbic acid or methylene anaerobic glycolysis for energy via the
blue 1)Embden-Meyerhof pathway
• Energy is needed for the main Embden-Meyerhof
2) Sulfhemoglobin pathway
• oxidation of Hgb by drugs or chemical • Maintain cation gradients
containing sulfur (inorganic sulfides ) ○ Keeps potassium inside and sodium outside the RBC
• Process is irreversible, so once Hgb has Maintain the RBC membrane flexibility
become sulfhemoglobin, it will remain for • Supply offshoot pathways
the life of the cell
• Unable to transport O2, leading to 2)Hexose monophosphate shunt
cyanosis. • Protection of the RBC from oxidant damage by
• Measured at 620nm production of reduced glutathione
• green-pigmented molecule
3) Methemoglobin reductase pathway
3) Carboxyhemoglobin • Maintaining iron in the ferrous form for Hgb to
• Hgb resulting from heme iron-binding limit the production of methemoglobin
carbon monoxide CO
• It has a 240 times high O2 affinity and 4) Rapoport-Luebering pathway
does not give up O2 to the tissues easily • Regulation of O2 delivery to tissues by the
production of 2,3 bisphosphoglycerate (2,3 BPG
Hemoglobin/Erythrocyte Breakdown Hemoglobin and Iron

1. Intravascular hemolysis (10%) 1. Most iron in the body is in hemoglobin and


a. Occurs when hemoglobin breaks down in the must be in the ferrous state (Fe2+) to be used.
blood and free hemoglobin is released into
plasma 2. Fe2+ binds to oxygen for transport to lungs and
body tissues.
b. Free hemoglobin binds to haptoglobin (major
free hemoglobin transport protein), hemopexin, 3.Ferric iron (Fe3+) is not able to bind to
and albumin, and it is phagocytized by liver hemoglobin, but does bind to transferrin.
macrophages.
4. Iron is an essential mineral and is not produced
c. Laboratory: by the body.
✓ Increased plasma hemoglobin
✓ serum bilirubin a. Serum iron measures the amount of Fe3+
✓ serum LD bound to transferrin.
✓ urine urobilinogen;
✓ hemoglobinuria and hemosiderinuria b. Total iron-binding capacity (TIBC) measures the
present total amount of iron that transferrin can bind
✓ decreased serum haptoglobin when fully saturated.

2. Extravascular hemolysis (90%) c. Serum ferritin is an indirect measurement of


storage iron in tissues and bone marrow.
a. Occurs when senescent/old RBCs are
phagocytized by macrophages in the liver or
spleen

b. Protoporphyrin ring metabolized to bilirubin


and urobilinogen; excreted in urine and feces

c. Globin chains are recycled into the amino acid


pool for protein synthesis.

d. Iron binds to transferrin and is transported to


bone marrow for new RBC production, or it is
stored for future use in the form of ferritin or
hemosiderin.
Hematology Stains 2. Nonvital monochrome stain
1. Nonvital (dead cell) polychrome stain
(Romanowsky) a. Stains specific cellular components
b. Prussian blue stain is an example.
a. Most commonly used routine peripheral blood 1) Contains potassium ferrocyanide, HC1, and a
smear stain safranin counterstain

b. Wright's stain contains 2) Used to visualize iron granules in RBCs


✓ methylene blue, a basic dye, which (siderotic iron granules), histiocytes, and urine
stains acidic cellular components (DNA epithelial cells
and RNA) blue
3. Supravital (living cell) monochrome stain
✓ eosin, an acidic dye, which stains basic
components (hemoglobin and a. Used to stain specific cellular components
eosinophilic cytoplasmic granules) red-
orange. b. No fixatives are used in the staining process.

c. Methanol fixative is used in the staining c. Includes:


process to fix the cells to the slide. 1) New methylene blue used to precipitate RNA
in reticulocytes;
d. Staining does not begin until a phosphate
buffer (pH between 6.4 and 6.8) is added 2) Neutral red with brilliant cresyl green as a
counterstain is used to visualize Heinz bodies;
Note: clinical disorders associated with Heinz bodies
✓ Causes of RBCs too red and WBC nuclei include G6PD deficiency and other unstable
poorly stained: Buffer or stain below pH hemoglobin disorders.
6.4, excess buffer, decreased staining
time, increased washing time, thin smear,
expired stains

✓ Causes of RBCs and WBC nuclei too blue:


Buffer or stain above pH 6.8, too little
buffer, increased staining time, poor
washing, thick smear, increased protein,
heparinized blood sample

Note:
✓ Examples of polychrome stains include:
Wright, Giemsa, Leishman, Jenner, May –
Grimwald
VI. Hemoglobin Measurements VII. Laboratory Evaluation of Blood
1) Cyanmethemoglobin method – is the
reference method . A) RBC indices

• It measures all form of hemoglobin 1. MCV (mean corpuscular volume):


except of sulfhemoglobin • it is an indicator of the average/mean
volume of erythrocytes (RBCs).
• It uses Drabkin’s Reagent –composition
• Lysing agent – its liberates hemoglobin • Reference range (Si/conventional units) is
• Potassium Ferricyanide – converts 80-100 femtoliters (fL),
hemoglobin to methemoglobin
• Potassium cyanide – forms the stable Formula : MCV (fL) = hct(%) x 10
pigment of cyanmethemoglobin RBC count (X 10^12/L)
• Read at 540nm

2)Geometric method 1. Increased in megaloblastic anemia,


• Principle : a given sample of blood can be hemolytic anemia with reticulocytosis,
equilibrated with oxygen under standard liver disease, and normal newborn
conditions of temp. And pressure
• Can measure only active hemoglobin b. Decreased in iron deficiency anemia ,
thalassemia , sideroblastic anemia and lead
3) Gravimetric method poising
• Determines hemoglobin concentration
via blood specific gravity 2. MCH (mean corpuscular hemoglobin):
• Uses copper sulfate solution (sp.gr: 1.053 • is an indicator of the average weight of
or 1.055) hemoglobin in individual RBCs
• Used for mass screening
• Cu sulfate is blue is color and turns green • Reference range (Si/conventional units)
with over used is 26-34 picograms (pg).
• RBC gravity = 1.053
FORMULA : MCH= Hemoglobin (g/dL) X10
4) Acid Hematin Methods RBC count (X 1012/L)
• Uses diluted hydrochloric acid 90.1N Hcl),
a. Increased in macrocytic anemia
to convert Hb to acid hematin which is
b.Decreased in microcytic hypochromic
then match with color standard in a
anemia
comparator block
• Uses sahli-hellige hemoglobinometer
upto 20ul
3) MCHC (mean corpuscular hemoglobin Formula
concentration): For automated cell counters using the MCV
• it is a measure of the average and RBC count:
concentration of hemoglobin in grams per Hct % = MCV (fL) X RBC count (X 10^12)
deciliter. 10
Using microhematocrit
• Reference range (conventional units) is 32- Hct %= packed RBC (mm) x 100
37 g/dL (SI units 320-370 g/L) Total volume (mm)

Formula : MCHC (g/dL) = Hb (g/dL) X 100 Increased Hct


Hct 1. Polycythemia Vera
2. Hemoconcentration
3. Dehydration
REMINDER 4. Megaloblastic anemia
a. 32-37 g/dL MCHC indicates normochromic RBCs. 5. High altitude

b. Lesser than (<) 32 g/dL MCHC indicates decreased Hct


hypochromic RBCs, which is seen in iron deficiency 1. Microcytic anemia
and thalassemia. 2. Excessive hydration
3. Menstruation
c. Greater than (>) 37 g/dL MCHC indicates a 4. Physiological hydremia of pregnancy
possible error in RBC or hemoglobin measurement,
or the presence of spherocytes. NOTE :
a. Spun microhematocrit is the reference
B) Hct (Hematocrit): aka Packed Red Cell Volume manual method.
• Hematocrit is the percentage of packed
RBCs in a given volume of whole blood. b. The buffy coat layer of leukocytes and
platelets, not included in the measurement,
• Reference range for hematocrit is age and can be seen between plasma (upper) and RBC
sex dependent (lower) layers.

• Reference range : C) Erythrocyte Sedimentation Rate (ESR)


Male (conventional units) is 41-53% (SI units
0.41-0.53 L/L) • Screening test used to screen or
Females (conventional units) is 36-46% (SI units monitor for various inflammatory
0.36-0.46 L/L). states.
• The ESR looks at how much RBC
settling will occur in a well-mixed
whole blood sample over a 1-hour
period
• RBCs normally have a net negative E) WBC count = cell counted x dilution factor x10
charge, causing them to repel each other Area counted
in a whole blood sample, leading to slow
settling of the RBCs over time Diluents for WBC : weak acid (1N HCl , Glacial
acetic acid ) , Turk’s diluting fluid etc
• When a change to the charge occurs
,usually resulting from increases in
plasma proteins, the cells become F) RDW (RBC distribution width): reflects the
attracted to each other, leading to degree of anisocytosis
increased settling speeds of the RBCs
Reference range (conventional units) is 11.5-
• Tests are performed with manual 14.5%.
Westergren and Wintrobe procedures or a. Determined from the RBC histogram
automated analyzers to allow for a faster
reading b. Increased proportional to the degree of
anisocytosis (variation in size); coefficient of
D) Manual RBC count using Hemacytometer variation of the mean corpuscular volume
• Dilutions are made using calibrated
pipettes and diluents c. High RDW: Seen post-transfusion, post-
treatment (e.g., iron, B12, or folic acid therapy),
• The same counting principle is applied to idiopathic sideroblastic anemia, in the presence
all blood cells expect with different of two concurrent deficiencies (iron and folic acid
diluents and counting area deficiencies

• Counting chamber used for manual cell G) Reticulocyte Count


counting, currently most frequently used for
body fluid cell counting • Manual counts are performed by
incubating (EDTA) whole blood with a
• Based on the use of a known counting area supravital stain, usually new methylene
of 9 squares each with an area of 1 mm2 and blue . If any RNA or residual organelles
a total volume of 0.9 mm3 for a total area of are present, they will take up the
9 mm3. A standard formula is used to supravital stain and are visible
determine a total cell count/m L (1 mm) microscopically.
• Various methods are used for the manual
Diluting fluids for RBC : reticulocyte count, including the Miller
Dacie’s or Formol citrate (best) , NSS ocular and other techniques to
determine the total percentage of
Formula for RBC count = reticulocytes present
Cell counted x Dilution Factor x 10 Formula for retic count=
Area (mm2) Number of reticulocyte x100
1000 RBC observed

RF: 0.5-1.5 %
VIII. Leukopoiesis 3. Myelocyte
• .The myeloid progenitor cell gives rise to a
committed progenitor cell that is acted on a. First stage where granulocyte types can
by growth factors to form granulocytes. be differentiated into eosinophils,
basophils, and neutrophil
• Classified as phagocytes (granulocytes,
monocytes) or immunocytes (lymphocytes, b. N:C ratio 2:1
plasma cells, and monocytes) c. Last stage capable of cell division
d. Round nucleus with coarse chromatin
• Granulocytes include neutrophils, e. Early myelocytes may have visible
eosinophils, and basophils. nucleoli.
f. Light blue to light pink cytoplasm
A) Maturation and Morphology of Immature g. Prominent golgi where (secondary)
Granulocytes granules, at first form there .
h. Cytoplasm formed a
1. Myeloblast: specific/secondary granules that
Earliest recognizable granulocyte precursor contain hydrolytic enzymes, including
Uncommitted when considering granulopoiesis alkaline phosphatase and lysozyme.

A) N:C ratio 7:1-4:1 I. Nonspecific/primary granules are


B) Round/oval nucleus with fine reddish-purple present and may still stain, With
staining chromatin successive mitoses, the number of
C) 2-5 nucleoli azurophilic granules (whose production
D)Dark blue cytoplasm has ceased at the end of the
E) No cytoplasmic granules promyelocyte stage) is diminished.
F)1 % of the nucleated cells in the bone marrow
j. Neutrophilic myelocyte makes up 13% of
2. Promyelocyte the nucleated cells in the bone marrow.
a. this stage encompasses the entire period
of production of azurophilic granules 4. Metamyelocyte
b. N:C ratio 3:1
c. Round/oval nucleus with slightly coarsening a. From this stage on, changes in the
chromatin cytoplasm are insignificant.
d.1-3 nucleoli b.Nucleus is indented in a kidney bean shape
e. Dark blue cytoplasm and has coarse, clumped chromatin.
f. Cytoplasm has large, nonspecific/primary c.Cytoplasm is pink and filled with pale blue to
granules containing myeloperoxidase. pink specific/secondary granules and tertiary
g. 2-5% of the nucleated cells in the bone granules are formed
marrow d.Nonspecific/primary granules are present but
usually do not stain.
e.Neutrophilic metamyelocyte makes up 16%
of the nucleated cells in the bone marrow.
5. Band neutrophil (stab form)
a. Nucleus is "C" or "S"-shaped with • Phagocytic cells present in the peripheral
coarse, clumped chromatin lacking circulation destroy foreign substances
segmentation. and microorganisms
• Constitute the majority of circulating
b. Cytoplasm is pink and filled with pale WBCs in adults
blue to pink specific/secondary granules.
f. Nonspecific/primary granules are • Normal neutrophil function (Phagocytosis
present but usually don't stain. Bactericidal activity)
○ Cells move to a site of inflammation, and
c. Band neutrophil makes up 12% of the granules are released to assist in travel and
nucleated cells in the bone marrow, and adhesion. Once at their target site, cells work to
0-5% of peripheral white blood cells eliminate the foreign material by phagocytosis
(WBCs). Granule contents and neutrophil extracellular
traps are used to trap and kill microorganism
d. Stored in the bone marrow and released
when there is an increased demand for
neutrophils • Location

B) Morphology of Mature Granulocytes ○ Present in a circulating pool in which


neutrophils travel throughout the peripheral
1) Segmented neutrophil (aka seg, circulation and
polymorphonuclear cell (PMN), and poly) ○ marginating pool in which neutrophils line the
walls of the vasculature, waiting to be called into
• Nucleus has coarse, clumped chromatin use.
with 3-5 lobes connected by thin ○ In the bone marrow, before release to the
filaments. peripheral vasculature, a storage pool and mitotic
• Cytoplasm is pink and filled with small, pool are present
pale blue to pink specific/ secondary
granules. Neutrophil Granules
• Nonspecific/primary granules are 1) Azurophilic granules (formed in
present but usually do not stain unless in promyelocyte stage)
response to infection or growth factor. • Lysosomal enzymes: acid
• Segmented neutrophil makes up 12% of hydrolases, acid phosphatase , β-
the nucleated cells in the bone marrow, glucuronidase Myeloperoxidase, Elastase,
and 50-80% of peripheral WBCs. Arylsulfatase & Cationic antibacterial
proteins
2)Specific granules (formed in myelocyte
stage) • Cells serve in immune regulation, from
• Lysozyme, Lactoferrin ,Collagenase antigen presentation to initiation of
Plasminogen activator &Aminopeptidase immune response

3) Tertiary granules 3. Basophil


• Gelatinase ,Cytoplasmic organelles &
Alkaline phosphatase Maturation is similar to eosinophil and neutrophil
maturation. Basophils have several different
types of granules.
2. Eosinophil
a.Cytoplasm contains large, purple-black,
Maturation is similar to neutrophil maturation, secondary granules that contain heparin and
although granule contents are different histamine.

a. Recognizable maturation stages include the b.Granules may be numerous and obscure the
eosinophilic myelocyte, eosinophilic nucleus, or they may "wash out" in staining
metamyelocyte, eosinophilic band, and (because the granules are water soluble) and
eosinophil (segmented form). leave empty areas in the cytoplasm.

b. Nucleus is usually bilobed. c. Basophils make up less than 0.1 % of the


nucleated cells in both the bone marrow and
c.Cytoplasm contains large, bright red-orange, peripheral blood
secondary granules that contain enzymes and
proteins. Function : In the blood only a few hours before
migrating to the site of inflammation in the tissue
d. Eosinophils make up less than 1% of the
nucleated cells in the bone marrow and 5% of • Cells have immunoglobulin E (IgE)
peripheral WBCs receptors that lead to their effectiveness
in allergic and hypersensitivity type I
Function : reactions. They also play a role in
a. In the blood only a few hours before seeking a initiating the immune response
tissue site such as nasal passages, skin, or urinary
tract • Basophils release a chemotactic factor
that attracts eosinophils to the site.
b. They can degranulate like neutrophils. They
express Fc receptors for IgE, which is a response
to parasitic infections (helimenths ) • NOTE: Mast cells are somewhat related
to basophils; however, they are tissue
c. They release substances that can neutralize cells used in allergic reactions and
products released by basophils and mast cells; inflammation
eosinophils modulate the allergic response.
IV. MONOCYTES AND MACROPHAGES
✓ Known as "scavenger cells" because of
• Monocytes form in the bone marrow, their ability to ingest foreign material
pass through the peripheral blood, and
then migrate into the tissues (macro- a. Blood monocytes ingest antigen-antibody
phages), where they fight infection. complexes and activated clotting factors,
limiting the coagulation response.
• Macrophages are named according to
their location in the body. b. Splenic macrophages remove old/damaged
RBCs and conserve iron for recycling.
✓ a. Monocytes —peripheral blood
✓ b. Kupffer cells—liver c. Liver macrophages remove fibrin
✓ c.Microglial cells—central nervous system degradation products.
✓ d. Osteoclasts—bone
✓ e. Langerhans' cells—skin d. Bone marrow macrophages remove
✓ f. Alveolar cells—lung abnormal RBCs, ingest bare megakaryocyte
nuclei or extruded RBC nuclei, and store and
• Functions supply iron for hemoglobin synthesis
✓ Cells are used in both innate and
adaptive immunity. They can recognize
and phagocytize foreign materials; in Monocyte Characteristics
addition they can serve as antigen
presenting cells to initiate T and B cells 1. Granules are lysosomes that contain hydrolytic
they are and can be used for house enzymes, including peroxidase and acid
keeping purposes to remove dead cells phosphatase.
and debris
2. Highly motile cell that marginates against
✓ They process ingested material and also vessel walls and into the tissues
process antigenic information, which is
relayed to the T-helper (CD4) 3. Reference range is 2-10% in peripheral blood.
lymphocyte. The T-helper lymphocyte
coordinates the immune response to Maturation and Morphology of Monocytes
foreign antigens.
1. Monoblast: Earliest recognizable monocyte
✓ They arrive at the site of inflammation precursor
after neutrophils. Unlike neutrophils, the a. 12-18 (Jim; N:C ratio 4:1
phagocytic process does not kill the
monocyte b. Round/oval eccentric nucleus with fine
chromatin; 1-2 nucleoli
✓ Very efficient phagocytic cells with
receptors for IgG or complement-coated c. Dark blue cytoplasm; may have a gray tint; no
organisms cytoplasmic granules
2. Promonocyte
a. N:C ratio 3:1 • The pluripotential stem cell gives rise to
b. Irregularly shaped, indented nucleus with fine the lymphoid progenitor cell that is acted
chromatin; 0-1 nucleoli on by colony stimulating
c. Blue to gray cytoplasm; fine azurophilic factors/interleukins/cytokines to form B
granules and T lymphocytes.

3. Monocyte • Pre-B lymphocytes differentiate in the
a. 12-20 bone marrow, and pre-T lymphocytes
differentiate in the thymus through
b. Horseshoe- or kidney-bean-shaped nucleus, antigen independent lymphopoiesis.
often with "brainlike" convolutions
• Bone marrow and thymus are primary
c. Fine, lacy chromatin lymphoid tissues.

d. Blue-gray cytoplasm; may have pseudopods • B- and T cells enter the blood and
and vacuoles populate the secondary lymphoid tissues
(lymph nodes, spleen, and Peyer's
e. Many fine azurophilic granules give the patches in the intestine), where antigen
appearance of "ground glass." contact occurs.

f. Transitional cell because it migrates into the Maturation and Morphology of Lymphocytes
tissue and becomes a fixed or free macrophage 1. Lymphoblast:
Earliest recognizable lymphocyte precursor
4. Macrophage: "Tissue monocyte"
a. 15-80 |xm a. 10-18 |xm;N:C ratio 4:1
b. Round/oval eccentric nucleus with fine
b. Indented, elongated, or egg-shaped nucleus chromatin; 1 or more nucleoli
with fine chromatin c. Dark blue cytoplasm; no cytoplasmic granules

c. Blue-gray cytoplasm with many vacuoles and 2. Prolymphocyte


coarse azurophilic granules; may contain ingested
material a. N:C ratio 3:1
b. Round or indented nucleus with coarsening
chromatin; 0-1 nucleoli
V. LYMPHOCYTES AND PLASMA CELLS c. Basophilic cytoplasm; no cytoplasmic granules
• Lymphocytes can be produced in both
the bone marrow and the lymphoid
tissues. Cells can return from an
inactive/resting form into active blasts, as
needed
3. Lymphocyte T lymphocyte function

a. Round, oval, or slightly indented nucleus; a. T cells provide cellular immunity. They are
condensed chromatin responsible for graft rejections and lysis of
b. Scant to moderate amount of blue cytoplasm; neoplastic cells, and they attack/destroy viral and
few azurophilic granules fungal organisms.
b. Obtain antigenic information from monocytes;
4. Reactive lymphocytes - have become activated this information is passed to other T cells and B
as part of the immune response. cells
c. Regulate humoral response by helping
✓ Associated with lymphocytosis and can antigens activate B cells
show the following characteristics:
d. End products of activation are
a. Generally, larger cell with increased cytokines/lymphokines/interleukins
amount of dark blue cytoplasm (RNA)
b. Fine chromatin pattern with nucleoli B Lymphocytes (B cells)
c. Irregular shape to the nucleus
d. Irregular shape to the cytoplasm (tags, 1. Become immunocompetent in the secondary
sharp ridges); indented by red cells lymphoid tissue; dependent on antigenic
stimulation.
a. Acquire specific receptors for antigens
T Lymphocytes (T cells) b. Make up 20% of the peripheral blood
lymphocytes
1. Become immunocompetent in the
secondary lymphoid tissue; dependent on 2. Identified by membrane markers CD19, CD20,
antigenic stimulation and others

a. Acquire specific receptors for antigens B lymphocyte function


b. Make up 80% of the peripheral blood
lymphocytes a. Contact with foreign antigens stimulates B
lymphocytes to become reactive lymphocytes,
2. They are identified by membrane markers with the characteristic morphology associated
CD2, CD3, and others. The markers appear, with reactivity.
disappear, and then reappear throughout cell
development b. Reactive lymphocytes transform into
immunoblasts, and then plasma cells that
produce antibodies to provide humoral
immunity.
c. Plasma cells • Maturation and selection of T cells occur
primarily in the thymus, and of B cells, in
1) End stage of B lymphocyte; dominant in lymph the marrow and peripheral lymphoid
nodes; not normally seen in circulation organs

2) Abundant blue cytoplasm with prominent • B cells develop in the bone marrow after
perinuclear (golgi) zone the hematopoietic stem cells have
3) Eccentric nucleus with a very coarse, clumped populated that organ. During adult life,
chromatin pattern generation of B cells occurs in the bone
4) Make up less than 4% of nucleated cells in the marrow.
bone marrow
- B cell differentiation can be divided
conveniently into two stages :
Natural Killer (NK)/l_arge Granular Lymphocytes
(LGLs) • The initial stage of B cell differentiation
involves the antigen-independent
1. Large cells with low N:C ratio, large cytoplasmic generation of diversity through
granules, and pale blue cytoplasm rearrangement of the Ig heavy and light
2. Lack B cell or T cell membrane markers; are chain genes.
CD16 and CD56 positive
3. Responsible for surveillance of cells for surface • The second stage is regulated by antigen
alterations such as tumor cells or cells infected triggering, T cell interaction,
with viruses macrophages, and various growth factors
4. Activated by IL-2 to express nonspecific . This stage occurs predominantly in the
cytotoxic functions secondary lymphoid organs
5. Attack antigens with attached IgG; called
antibody-dependent cytotoxic cells SECONDARY LYMPHOID TISSUE

• late fetal and postnatal life, lymphocytes


IMPORTANT are produced in the secondary lymphoid
• Primary lymphoid organs are : bone (B tissue: spleen, lymph nodes, and
cell development ) marrow and thymus intestine , MALT , peyer patches
(T-cell development )
• lymphocyte precursors originate in the • The secondary lymphoid organs are
bone marrow and undergo antigen- composed of a mixture of B cells and T
independent lineage commitment cells.
• Lymphopoiesis in secondary lymphoid
organs depends solely on antigenic
stimulation
II.ERYTHROCYTIC DISORDERS

A) Erythrocytic Morphology and Associated 5. Poikilocytosis


Disease (Size and Shape)
a. General term to describe variation in shape
1. Normocytes (discocytes) are normal b. Associated with a variety of pathologic
erythrocytes that are approximately the same conditions
size as the nucleus of a small lymphocyte
6. Echinocytes - crenated and burr cells
2. Macrocytes
a. RBCs greater than 8 micro m in diameter; a. Have evenly spaced round projections; central
MCV greater than 100 fL pallor area present

b. Seen in megaloblastic anemias, such as b. Caused by changes in osmotic pressure


B12/folate deficiency
b. Seen in liver disease, uremia, heparin therapy,
c. Seen in non-megaloblastic anemia of liver pyruvate kinase deficiency, or as artifact
disease or accelerated erythropoiesis; also
seen in normal newborns 7. Acanthocytes (spur cells)

3. Microcytes a. Have unevenly spaced pointed projections;


a. RBCs less than 6 ixm in diameter; MCV lack a central pallor area
less than 80 fL
b. Seen in iron-deficiency anemia, b. Associated with alcoholic liver disease, post-
thalassemia, sideroblastic anemia, and splenectomy, and abetalipoproteinemia
anemia of chronic disease (Neuroacanthocytosis, McLeod’s syndrome)

4. Anisocytosis c. Caused by excessive cholesterol in the


a. Variation in RBC size volume or diameter, membrane
indicating a heterogeneous RBC population
(dimorphism) 8. Target cells (codocytes or Mexican hat cells)

b. Correlates with RDW (red blood cell a. Show a central area of hemoglobin
distribution width), especially when the surrounded by a colorless ring and a peripheral
RDW exceeds 15.0% ring of hemoglobin; cells have an increased
surface-to-volume ratio
c. Seen post-transfusion, post-treatment for
a deficiency (e.g., iron), presence of two b. Seen in liver disease, hemoglobinopathies,
concurrent deficiencies (e.g., iron and thalassemia, iron-deficiency anemia
vitamin B12), hemolytic and idiopathic
sideroblastic anemia c. Caused by excessive cholesterol in the
membrane or a hemoglobin distribution
imbalance
9. Spherocytes
12. Helmet cells (horn cells or keratocytes)
a. Disk-shaped cell with a smaller volume than a
normal erythrocyte; cells have a decreased a. Interior portion of cell is hollow, rbc fragment
surface-to-volume ratio resembling a horn or helmet
b. Seen in microangiopathic hemolytic anemias
b. Lack a central pallor area
c. Associated with defects of the red cell 13. Schistocytes (RBC fragments)
membrane proteins
d. MCHC may be >37%; increased osmotic a. Damaged RBC; Fragmented RBC resulting
fragility from rupture in the peripheral circulation

e. Damaged RBC; seen In b. Seen in


✓ hereditary spherocytosis ✓ microangiopathic hemolytic anemias
✓ G6PD deficiency, (e.g., DIC, HUS, TTP),
✓ immune hemolytic anemias ✓ thermal injury,
✓ renal transplant rejection
f. Microspherocytes (<4 |jim) are frequently ✓ G6PD deficiency
seen in severe thermal injury (burns) (along with ✓ Traumatic cardiac hemolysis
schistocytes) ✓ Extensive burns (along with
microspherocytes)
10. Teardrops (dacryocytes)
14. Stomatocytes (mouth cells)
a. Pear-shaped cell with one blunt projection
b. Seen in a. Characterized by an elongated or slit-like area
✓ megaloblastic anemias, of central pallor
✓ Thalassemia b. Seen in
✓ extramedullary hematopoiesis ✓ liver disease
(myelofibrosis, myelophthisic anemia) ✓ hereditary stomatocytosis
✓ artifact
11. Sickle cells (drepanocytes) ✓ Rh deficiency syndrome
✓ Acquired stomatocytosis
a. Shapes vary but show thin, elongated, (liverdisease,alcoholism)
pointed ends and will appear crescent shaped; c. Caused by osmotic changes due to cation
usually lack a central pallor area imbalance (Na+/K+)

b. Contain polymers of abnormal hemoglobin S 15. Elliptocytes (ovalocytes)


c. Seen in hemoglobinopathies SS, SC, SD, and a. Cigar- to egg-shaped erythrocytes
sickle cell thalassemia b. Associated with defects of the red cell
membrane protein
d. Cell shape is caused by cell membrane
alterations due to an amino acid

2. seen in 4. Pappenheimer bodies


✓ hereditary elliptocytosis,
✓ iron-deficiency anemia (pencil forms) a. Small, irregular, dark-staining iron granules
✓ megaloblastic anemia (macro- usually clumped together at periphery of the cell
ovalocytes)
✓ thalassemia major b. Stain with Perl's Prussian blue stain; appear
dark violet with Wright's stain
B. Erythrocyte Inclusions and Associated
Diseases c. Caused by an accumulation of ribosomes,
mitochondria, and iron fragments
1.Howell-Jolly bodies d. Seen in
a. Small, round DNA fragments (0.5-1.0 jxm in ✓ sideroblastic anemia
diameter) usually one per cell, but can be ✓ Hemoglobinopathies
multiple ✓ Thalassemia
✓ megaloblastic anemia
b. Stain dark purple to black with Wright's stain ✓ Hyposplenism
c. Not seen in normal erythrocytes; normally ✓ myelodysplastic syndrome (RARS)
pitted by splenic macrophages
5. Cabot rings
d. Seen in a. Thin, red-violet, single to multiple ring like
✓ sickle cell anemia structures that may appear in loop or figure-
✓ beta-thalassemia major eight shapes
✓ severe hemolytic anemias
✓ megaloblastic anemia, b. Seen in
✓ alcoholism, ✓ megaloblastic anemia
✓ post-splenectomy ✓ myelodysplastic syndromes
✓ lead poisoning
3. Basophilic stippling c. Composed of fragments of nuclear
a. Multiple, tiny, fine, or coarse inclusions material(Remnant of mitotic spindle)
(ribosomal RNA remnants) evenly dispersed
throughout the cell; "blueberry bagel" 6. Hemoglobin C crystals
appearance
b. Stain dark blue with Wright's stain a. Condensed, intracellular, rod-shaped crystal.
c. Seen in Hexagonal crystal of dense Hgb formed within
✓ Thalassemia the RBC membrane
✓ megaloblastic anemias
✓ sideroblastic anemia b. Seen in
✓ lead poisoning ✓ hemoglobin C or SC disease, but not in
✓ Alcoholism trait
✓ Hemoglobinopathies
✓ Abnormal heme synthesis

7. Hemoglobin SC crystals (Washington


monument) d. Seen in
a. 1-2 blunt, fingerlike projections extending ✓ iron-deficiency anemia
from the red cell membrane ✓ Thalassemias
✓ anemia of chronic disease
b. Seen in hemoglobin SC disease ✓ sideroblastic anemia
✓ myelodysplastic syndromes
8. Heinz bodies
3. Polychromasia
a. Multiple inclusions ranging in size from 0.3 to a. Variation in hemoglobin content showing a
2.0 slight blue tinge when stained with Wright's
stain; residual RNA
b. Invisible with Wright's stain; must use a
supravital stain to visualize b. Indicates reticulocytosis; supravital
reticulocyte stain to enumerate
c. Seen in
✓ G6PD deficiency c. Usually slightly macrocytic
✓ beta-thalassemia major,
✓ Hgb H disease 4. Hyperchromasia (term no longer used)
✓ unstable hemoglobinopathies
✓ drug-induced anemias a. Current terminology is spherocyte; lacks a
d. Represent denatured hemoglobin central pallor area

9. Malarial parasites I. Abnormal Erythrocyte Distributions and


✓ include P. vivax, P. falciparum, P. Associated Diseases
malariae and P. ovale
1. Rouleaux
H. Erythrocyte Hemoglobin Content and a. Stacking or "coining" pattern of erythrocytes
Associated Diseases due to abnormal or increased plasma proteins

1. Normochromasia: Cells have the normal one- b. May see excessively blue color to smear
third clear, central pallor area macroscopically and microscopically

2. Hypochromasia c. Seen in
✓ Hyperproteinemia
a. Central pallor area is greater than one-third ✓ multiple myeloma
the diameter of the cell ✓ Waldenstrom macroglobulinemia
b. MCH and MCHC usually decreased ✓ conditions that produce increased
c. Often associated with microcytosis fibrinogen (chronic inflammation)
d. May be artifact; considered normal in thicker
area of the peripheral smear

e. True rouleaux formation is determined in the


thin area of the peripheral smear.

. Agglutination
a. Characterized by clumping of erythrocytes with no pattern

b. Occurs when erythrocytes are coated with IgM antibodies and complement

c. Seen in

✓ cold autoimmune hemolytic anemia (cold agglutinin disease)

d. Warm blood to 37°C to correct a false low RBC and hematocrit, and false high MCHC (>37 g/dL)
when using an automated cell counting instrument

IX. ANEMIA AND RED BLOOD CELL DISORDERS


Introduction • Common tests for initial anemia
• Anemia is defined as a decrease in evaluation
erythrocytes and hemoglobin,
resulting in decreased oxygen delivery ✓ Complete blood count (CBC) with
to the tissues. peripheral smear review

• The anemias can be classified ✓ Smear examination will reveal the


morphologically using RBC indices appearance of RBCs (anisocytosis and
(MCV, MCH, and MCHC). poikilocytosis and inclusions)

• They can also be classified based on ✓ Reticulocyte count ,Shows the bone
etiology/cause. marrow response to decreases in
• Anemia is suspected when the RBCs
hemoglobin is <12 g/dL in men or <11
g/dL in women. -Anemias may be classified by
combinations of different criteria
-The cause of Anemia fall into three major
pathophysiological categories : 1.Morphology -RBC indices are used to
gauge size and hemoglobinization
✓ Blood loss ( acute or chronic )
✓ Impaired red cell production ✓ Normocytic/normochromic
✓ Accelerated red cell destruction ✓ Microcytic/hypochromic
✓ Macrocytic/normochromic
2. Function -Defects leading to RBC 2. Anemia of chronic disease (ACD)
decreases
✓ Proliferation: RBCs are not produced - Adequate iron stores that have impaired
at normal rates release for incorporation into heme/RBCs
✓ Maturation: RBCs are produced in
the marrow but may not mature a. Due to an inability to use available iron for
appropriately hemoglobin production
✓ Survival: RBCs are produced
appropriately but are lost/destroyed b. Impaired release of storage iron associated
prematurely with increased hepcidin levels

1) Hepcidin is a liver hormone and a positive


A. Impaired or Defective Production acute-phase reactant. It plays a major role in
Anemias body iron regulation by influencing intestinal
I.IRON AND HEME DISORDERS iron absorption and release of storage iron
1. Iron-deficiency anemia - Lack of iron to from macrophages.
make adequate heme
2) Inflammation and infection cause
a. Most common form of anemia in the hepcidin levels to increase; this decreases
United States release of iron from stores.

b. Prevalent in infants and children, c. Laboratory:


pregnancy, excessive menstrual flow, elderly ✓ Normocytic/normochromic anemia, or
with poor diets, malabsorption syndromes, slightly microcytic/hypochromic
chronic blood loss (GI blood loss, hookworm anemia
infection) ✓ increased ESR
✓ normal to increased ferritin
c. Laboratory: ✓ low serum iron and TIBC
✓ Microcytic/hypochromic anemia
✓ serum iron, ferritin, d. Associated with
hemoglobin/hematocrit, RBC indices, ✓ persistent infections
and reticulocyte count low ✓ chronic inflammatory disorders (SLE,
✓ RDW and total iron-binding capacity rheumatoid arthritis, Hodgkin
(TIBC) high; smear shows lymphorna, cancer)
ovalocytes/pencil forms
e. Anemia of chronic disease is second only to
d. Clinical symptoms: Fatigue, dizziness, iron deficiency as a common cause of anemia
pica, stomatitis (cracks in the corners of the
mouth), glossitis (sore tongue), and
koilonychia (spooning of the nails)
3. Sideroblastic anemia 2) Secondary—reversible; causes include :
-Adequate/excess iron that is not able to be
effectively incorporated into heme ✓ alcohol
✓ anti-tuberculosis drugs
a. Caused by blocks in the protoporphyrin ✓ Chloramphenicol
pathway resulting in defective hemoglobin ✓ Lead toxin
synthesis and iron overload
f. Laboratory:
b. Excess iron accumulates in the ✓ Microcytic/hypochromic anemia
mitochondrial region of the immature ✓ increased ferritin and serum iron
erythrocyte in the bone marrow and ✓ TIBC is decreased
encircles the nucleus; cells are called ringed
sideroblasts.
4-Hemochromatosis
c. Excess iron accumulates in the
mitochondrial region of the mature • Iron problem that does not involve
erythrocyte in circulation; cells are called anemia • Increased iron stores
siderocytes; inclusions are siderotic granules (absorption greater than loss)
(Pappenheimer bodies on Wright's stained • Stored as ferritin and hemosiderin
smears). o Often stored around organs
(heart, liver, pancreas)
d. Siderocytes are best demonstrated using
Perl's Prussian blue stain. -Acquired
✓ Transfusion related
e. Two types of sideroblastic anemia: ○ In cases of chronic transfusion, the
body recycles the iron from transfused RBCs,
1) Primary—irreversible; cause of the blocks in addition to its own senescent RBCs
unknown ✓ Chronic liver disease
✓ Alcoholism
a) Two RBC populations (dimorphic) are seen. ✓ Supplemental or dietary iron
b) This is one of the myelodysplastic overload
syndromes—refractory anemia with ringed -Inherited
sideroblasts (RARS) (Aquired ) • Several known mutations
✓ Classic hereditary
C) Hereditary - X-Linked or autosomal hemochromatosis, associated with
the HFE gene
✓ Hepcidin mutations, associated with
the HAMP gene
5-Lead poisoning Note :
The nucleus matures slower than the
a. Multiple blocks in the protoporphyrin cytoplasm (asynchronism). Megaloblastic
pathway affect heme synthesis maturation is seen.
b. Seen mostly in children exposed to lead-
based pain b. Caused by either a vitamin B12 or folic
c. Clinical symptoms: Abdominal pain, muscle acid deficiency
weakness, and a gum lead line that forms
from blue/black deposits of lead sulfate c. Laboratory:
d. Laboratory: ✓ Fancytopenia
✓ Normocytic/normochromic anemia ✓ macrocytic/normochromic anemia
with characteristic coarse basophilic with oval macrocytes and teardrops
stippling ✓ hypersegmented neutrophils
✓ inclusions include Howell-Jolly bodies
6. Porphyrias ✓ nucleated RBCs
✓ basophilic stippling
a. These are a group of inherited disorders ✓ Pappenheimer bodies
characterized by a block in the ✓ Cabot rings iron levels due to
protoporphyrin pathway of heme synthesis. destruction of fragile, megaloblastic
Note : Heme precursors before the block cells in the blood and bone marrow
accumulate in the tissues, and large amounts
are excreted in urine and/or feces. d. Vitamin B12 deficiency (cobalamin)

b. Clinical symptoms: Photosensitivity, Causes for vitamin B12 deficiency


abdominal pain, CNS disorders ✓ Poor diet
c. Hematologic findings are insignificant ✓ Increases in need ( Pregnancy ○
Lactation ○ Growing children)
II. MACROCYTIC ANEMIAS ✓ Impaired absorption and Inability to
obtain vitamin B12 from food in the
-There are 2 types of anemia under the stomach
macrocytic anemias : ✓ Competition for vitamin B12
✓ Megaloblastic anemia (Diphyllobothrium latum , Intestinal
✓ Nonmegaloblastic macrocytic anemia bacteria in blind loop syndrome
✓ A total vegetarian diet
A) Megaloblastic anemias

a. Defective DNA synthesis causes abnormal


nuclear maturation; RNA synthesis is normal,
so the cytoplasm is not affected.
✓ The Schilling test is no longer
1)Intrinsic factor is secreted by parietal cells performed regularly in the United
and is needed to bind vitamin B12 for States. Homocysteine and
absorption into the intestine. Methylmalonic acid (MMA) are
replacing this test, because they are
a) Pernicious anemia: better indicators of the deficiency.
Caused by
✓ deficiency of intrinsic factor, e. Folic acid deficiency
✓ antibodies to intrinsic factor
✓ or antibodies to parietal cells ✓ causes a megaloblastic anemia with a
✓ Autoimmune disease blood picture and clinical symptoms
✓ Helicobacter pylori infection similar to vitamin B12 deficiency
✓ gastrectomy except there is no CNS involvement.

B)Characterized by achlorhydria and atrophy ✓ It is associated with poor diet,


of gastric parietal cells pregnancy, or chemotherapeutic anti-
folic acid drugs such as methotrexate.
2) Clinical symptoms: Jaundice, weakness, ✓ Excessive loss , May occur in renal
sore tongue (glossitis), and gastrointestinal dialysis patients, so patients are
(GI) disorder, numbness and other CNS supplemented with folic acid
problems .
✓ Folic acid has low body stores.
3) Vitamin B12 deficiency takes 3-6 years to
develop because of high body stores B) Non-megaloblastic macrocytic anemias
✓ Macrocytosis without megaloblastic
Note : changes can occur.
✓ In vitamin B12 deficiencies, ✓ The erythrocytes are round, not oval
prolonged/severe cases may have as is seen in the megaloblastic
demyelination of the neurons; folic anemias.
acid deficiency does not have ✓ They results from other causes
neurologic involvement • Liver disease
• Alcoholism
✓ Schilling test - Classic two-part test • Hypothyroidism
used to determine if the cause of • Reticulocytosis
vitaminB12 deficiency is Note :
malabsorption, dietary deficiency, or They are Normal in babies , After delivery,
a lack of intrinsic factor macrocytosis and reticulocytosis are normal
III. HYPOPROLIFERATIVE DISORDERs ✓ Patients have a poor prognosis with
complications that include bleeding,
-Disorders occurring as a result of decreased infection, and iron overload due to
or absent production of hematopoietic cells frequent transfusion needs
in the bone marrow . This include
✓ Treatment includes bone marrow or
✓ Aplastic anemia, both inherited and stem cell transplant and
acquired immunosuppression.
✓ Other disorders resulting from ✓ Can be genetic, acquired, or
decreases in bone marrow production idiopathic
include
1) Genetic aplastic anemia (Fanconi
▫ Pure red cell aplasia anemia)
▫ Congenital dyserythropoietic anemia a) Autosomal recessive trait
▫ Myelophthisic anemia b) Dwarfism, renal disease, mental
▫ Anemia resulting from chronic kidney retardation
disease
c) Strong association with malignancy
1)Aplastic anemia development, especially acute
lymphoblastic leukemia
✓ Rare disorders characterized by
pancytopenia in the peripheral 2) Acquired aplastic anemia (secondary)
circulation. a. Bone marrow failure caused by:
causes pancytopenia
a) Antibiotics: Chloramphenicol and
✓ Result from decreased bone marrow sulfonamides
production of RBC, WBC, and platelets
because of deficiency or damage of b) Chemicals: Benzene and herbicides
hematopoietic stem cells
Laboratory: c) About 30% of acquired aplastic
▪ Decrease in hemoglobin/hematocrit anemias are due to drug exposure.
and reticulocytes;
normocytic/normochromic anemia d) Viruses: B19 parvovirus secondary to
▪ no response to erythropoietin hepatitis, measles, CMV, and Epstein-Barr
virus
✓ Most commonly affects people e) Radiation or chemotherapy
around the age of 50 and above. It
can occur in children.
f) Myelodysplastic syndromes, leukemia, 3)Myelophthisic (marrow replacement)
solid tumors, paroxysmal nocturnal anemia
hemoglobinuria
a. Hypoproliferative anemia caused by
3) Idiopathic (primary): replacement of bone marrow hematopoietic
✓ 50-70% of aplastic anemias have no cells by malignant cells or fibrotic tissue
known cause.
b. Associated with cancers (breast, prostate,
2) Pure Red Cell Aplasia lung, melanoma) with bone metastasis
- leukocytes and platelets normal in
number ✓ c. Laboratory:

✓ Bone marrow exhibits decreased Normocytic/normochromic anemia;


production of RBCs and RBC leukoerythroblastic blood picture
precursors, whereas other cell lines
are present and produced normally

✓ seen in children, often related to viral


infection ,Treated by transfusion, as
needed, although most patients
eventually restore their ability to
produce RBCs

✓ Congenital

• Diamond-Blackfan anemia -
Mutations are usually autosomal
dominant; however, they also may occur
sporadically
✓ IV.Blood Loss Anemia V. Hemolytic Anemias Due to Intrinsic
Defects
✓ 1. Acute blood loss anemia
• Hemolytic anemia can be due to
✓ a. Characterized by a sudden loss of intrinsic factors, usually inherited,
blood resulting from trauma or other such as disorders of the red cell
severe forms of injury membrane or enzymes, or
hemoglobinopathy.
✓ b. Clinical symptoms: Hypovolemia,
rapid pulse, low blood pressure, pallor 1. All cause a normocytic/normochromic
anemia; usually hereditary with
✓ c. Laboratory: reticulocytosis due to accelerated
✓ Normocytic/normochromic anemia destruction

✓ initially normal reticulocyte count 2. Hereditary spherocytosis


hemoglobin/hematocrit
a. Most common membrane defect;
✓ in a few hours, increase in platelet autosomal dominant; characterized by
count and leukocytosis with a left splenomegaly, variable degree of anemia,
shift, drop in hemoglobin/hematocrit spherocytes on the peripheral blood
and RBC; reticulocytosis in 3-5 days smear

2. Chronic blood loss anemia b. Increased permeability of the


membrane to sodium , Results in loss of
a. Characterized by a gradual, long-term loss membrane fragments
of blood; often caused by gastrointestinal
bleeding c. Erythrocytes have decreased surface
area-to-volume ratio; rigid spherocytes
b. Laboratory: culled/removed by splenic macrophages

✓ Initially normocytic/normochromic d. Laboratory:


anemia that over time causes a ✓ Spherocytes
decrease in hemoglobin/hematocrit; ✓ MCHC may be >37 g/dL
gradual loss of iron causes ✓ increased osmotic fragility
microcytic/hypochromic anemia ✓ increased serum bilirubin
3. Hereditary elliptocytosis (ovalocytosis)

a. Autosomal dominant; most persons


asymptomatic due to normal erythrocyte b. Increased cholesterol:lecithin ratio in
life span; >25% ovalocytes on the the membrane due to abnormal plasma
peripheral blood smear lipid concentrations; absence of serum p-
lipoprotein needed for lipid transport
b. Membrane defect is caused by
polarization of cholesterol at the ends of
the cell rather than around pallor area. 6. G6PD (glucose-6-phosphate
dehydrogenase) deficiency
4. Hereditary stomatocytosis
a. Sex-linked enzyme defect; most
a. Autosomal dominant; variable degree common enzyme deficiency in the hexose
of anemia; up to 50% stomatocytes on monophosphate shunt
the blood smear
b. Reduced glutathione levels are not
b. Membrane defect due to abnormal maintained because of decreased NADPH
permeability to both sodium and generation.
potassium; causes erythrocyte swelling
c. Results in oxidation of hemoglobin to
5. Hereditary acanthocytosis methemoglobin (Fe3+); denatures to
(abetalipoproteinemia) form Heinz bodies

a. Autosomal recessive; mild anemia d. Usually, not anemic until oxidatively


associated with steatorrhea, neurological challenged (primaquine, sulfa drugs);
and retinal abnormalities then severe hemolytic anemia with
reticulocytosis

NOTE :50-100% of erythrocytes are


acanthocytes
✓ Ham's and sugar water tests used in
7. Pyruvate kinase (PK) deficiency diagnosis

a. Autosomal recessive; most common ✓ increased incidence of acute leukemia


enzyme deficiency in Embden Meyerhof
pathway NOTE:
Although Ham's and sugar water tests have
b. Lack of ATP causes impairment of the been traditionally used in diagnosis of PNH,
cation pump that controls intracellular the standard now used is flow cytometry to
sodium and potassium levels. detect deficiencies for surface expression of
glycosyl phosphatidylinositol (GPI)-linked
c. Decreased erythrocyte deformability proteins such as CD55 and CD59.
reduces their life span.

d. Severe hemolytic anemia with VI. Hemolytic Anemias Due to


reticulocytosis and echinocytes Extrinsic/Immune Defects

8. Paroxysmal nocturnal hemoglobinuria -Hemolytic anemia may be caused by


extrinsic factors, usually acquired, such as
a. An acquired membrane defect in which chemical agents or antibodies
the red cell membrane has an increased
sensitivity for complement binding as 1. All cause a normocytic/normochromic
compared to normal erythrocytes anemia due to defects extrinsic to the RBC.
All are acquired disorders that cause
b. Etiology unknown accelerated destruction with reticulocytosis.

c. All cells are abnormally sensitive to lysis by 2. Warm autoimmune hemolytic anemia
complement. (WAIHA)

d. Characterized by: a. RBCs are coated with IgG and/or


✓ Pancytopenia complement. Macrophages may phagocytize
✓ chronic intravascular hemolysis these RBCs, or they may remove the antibody
causes hemoglobinuria and or complement from the RBC's surface,
hemosiderinuria at an acid pH at causing membrane loss and spherocytes.
night.
✓ PNH noted for low leukocyte alkaline
phosphatase (LAP) score;

✓ increased bilirubin, reticulocyte count


b. 60% of cases are idiopathic ✓ positive DAT detects complement-
coated RBCs
c.other cases are secondary to diseases that
alter the immune response (e.g., chronic d. If antibody titer is high enough, sample
lymphocytic leukemia, lymphoma); can also must be warmed to 37°C to obtain accurate
be drug induced. RBC and indices results.

c. Laboratory:
✓ Spherocytes, MCHC may be >37 g/dL 4. Paroxysmal cold hemoglobinuria (PCH)
✓ increased osmotic fragility& bilirubin
✓ reticulocyte count; occasional nRBCs a. An IgG biphasic Donath-Landsteiner
present antibody with P specificity fixes complement
✓ positive direct antiglobulin test (DAT) to RBCs in the cold (less than 20°C); the
helpful in differentiating from complement-coated RBCs lyse when warmed
hereditary spherocytosis. to 37°C

3. Cold autoimmune hemolytic anemia b. Can be idiopathic, or secondary to viral


(CAIHA or cold hemagglutinin disease) infections (e.g., measles, mumps) and non-
Hodgkin lymphoma
a. RBCs are coated with IgM and complement
at temperatures below 37°C. RBCs are lysed c. Laboratory
by complement or phagocytized by ✓ Variable anemia following hemolytic
macrophages. process
✓ increased bilirubin and plasma
Note: Antibody is usually anti-I but can be hemoglobin
anti-i. ✓ decreased haptoglobin

b. Can be idiopathic, or secondary to ✓ DAT may be positive; Donath-


Mycoplasma pneumoniae, lymphoma, or Landsteiner test positive
infectious mononucleosis

c. Laboratory:
✓ Seasonal symptoms; RBC clumping
can be seen both macroscopically and
microscopically
✓ MCHC >37 g/dL
VII. Hemolytic Anemias Due to c. Thrombotic thrombocytopenic purpura
Extrinsic/Non-lmmune Defects (TTP)

1. All cause a normocytic/normochromic 1) TTP occurs most often in adults.


anemia caused by trauma to the RBC.
2) It is likely due to a deficiency of the
Note:All are acquired disorders that cause enzyme ADAMTS 13 that is responsible for
intravascular hemolysis with schistocytes and breaking down large von Willebrand factor
thrombocytopenia. . multimers. When multimers are not broken
2. Microangiopathic hemolytic anemias down, clots form, causing RBC fragmentation
(MAHAs) and central nervous system impairment

a.Disseminated intravascular coagulation 3. March hemoglobinuria:


(DIC)
✓ Transient hemolytic anemia that
1) Systemic clotting is initiated by activation occurs after forceful contact of the
of the coagulation cascade due to toxins or body with hard surfaces (e.g.,
conditions that trigger release of marathon runners, tennis players)
procoagulants (tissue factor). Multiple organ
failure can occur due to clotting. 4. Other causes

2) Fibrin is deposited in small vessels, causing a. Infectious agents (e.g., P. falcipamm,


RBC fragmentation. Clostridium perfringens) damage the RBC
membrane. Schistocytes and spherocytes are
seen on the blood smear.
b. Hemolytic uremic syndrome (HUS)
b. Mechanical trauma, caused by prosthetic
1) Occurs most often in children following a heart valves (Waring blender syndrome),
gastrointestinal infection (e.g., E. coli) chemicals, drugs, and snake venom, damage
the RBCs through various mechanisms.
2) Clots form, causing renal damage
c. Thermal burns (third degree) cause direct
. damage to the RBC membrane, producing
acute hemolysis, which is characterized by
severe anemia with many schistocytes and
micro-spherocytes.
V. HEMOGLOBINOPATHIES
Introduction ✓ It results in a decrease in hemoglobin
✓ Disorders resulting from genetic solubility and function. Defect is
mutations that lead to structural inherited from both parents
changes in the Hgb molecule
2. No Hgb A is produced, and approximately
✓ Most occur because of amino acid 80% Hgb S and 20% Hgb F (the compensatory
substitutions in the b-globin chains hemoglobin) are seen. Hgb A2 is variable.
(qualitative defect)
3. Hemoglobin insolubility results when
✓ Numerous Hgb variants exist; deoxyhemoglobin is formed. Hemoglobin
however, several are more common crystallizes in erythrocytes. It is
than others characterized by the classic sickled shape of
erythrocytes
✓ Homozygous/disease conditions
(both globin chains affected) are more 3. Diagnosis is made after 6 months of age
serious than heterozygous/trait (time of beta-gamma globin chain switch),
conditions (only one globin chain with life expectancy of 50 years with proper
affected). treatment. Death usually results from
infection or congestive heart failure.
✓ Target cells are associated with the
hemoglobinopathies. 4. Laboratory :

✓ Hemoglobin electrophoresis, ✓ Severe normochromic/normocytic


isoelectric focusing and/or DNA (PCR) hemolytic anemia with
analysis may be used to confirm the polychromasia resulting from
diagnosis premature release of reticulocytes

✓ The amino acid substitution causing ✓ bone marrow erythroid hyperplasia


formation of Hgb S is the most (M:E ratio decreases)
common, Hgb C is the second most
common, and Hgb E is the third most ✓ Sickle cells, target cells, nucleated
common. RBCs, Pappenheimer bodies, and
Howell-Jolly bodies are seen
A) Sickle Cell Disease (Hgb SS)
-Homozygous for Hgb S ✓ Increased bilirubin and decreased
1. Sickle cell disease is caused when haptoglobin are characteristic due to
valine replaces glutamic acid at position hemolysis.
6 on both beta chains.
✓ Hgb S migrates with hemoglobins D 3. No Hgb A is produced; approximately 90%
and G on alkaline hemoglobin Hgb C , 2% Hgb A2, and 7% Hgb F are
electrophoresis; can differentiate produced. Mild anemia may be present.
using acid electrophoresis
4. Hgb C migrates with hemoglobins A2, E,
✓ Positive hemoglobin solubility and O on alkaline hemoglobin electrophresis;
screening test can differentiate hemoglobins using acid
electrophoresis
B) Sickle Cell Trait (Hgb AS)
5.The heterozygous Hgb C trait patient is
-Heterozygous for Hgb S ,Majority of Hgb asymptomatic, with no anemia; the one
present is Hgb A normal beta chain is able to produce
approximately 60% Hgb A and 40% Hgb C,
1- Sickle cell trait is caused when valine with normal amounts of Hgb A2 and Hgb F.
replaces glutamic acid at position 6 on one
beta chain. Defect is inherited from one 6.Solubility testing is negative
parent. One normal beta chain can produce
some Hgb A. 7.Laboratory:

2. Approximately 60% Hgb A and 40% Hgb S ✓ Normochromic/normocytic anemia


are produced, with normal amounts of Hgbs with target cells; characterized by
A2 and F. intracellular rod like C crystals

3. Sickle cell trait generally produces no Note : Solubility Test for Hemoglobin S (Sickle
clinical symptoms. Anemia is rare but, if Cell Prep)
present, will be normochromic/normocytic, 1. Hemoglobin S is insoluble when combined
and sickling can occur during rare crisis states with a reducing agent (sodium dithionite).
of hypoxia (same as in Hgb SS).
2. Hgb S will crystallize and give a turbid
4. Positive hemoglobin solubility screening appearance to the solution.
test
5. Apparent immunity to Plasmodium 3.The test will not differentiate homozygous
falciparum from heterozygous conditions containing Hgb
S.
C) Hemoglobin C Disease/Hgb CC
4. Follow up a positive solubility test with
1. Hgb C disease is caused when lysine hemoglobin electrophoresis
replaces glutamic acid at position 6 on both
beta chains. Defect is inherited from both
parents.
D. Hgb SC Disease d. Hgb E migrates with hemoglobins A2, C,
and O on alkaline hemoglobin
1. Hgb SC disease is a double heterozygous electrophoresis
condition where an abnormal sickle gene
from one parent and an abnormal C gene
from the other parent is inherited. Note : Solubility testing is negative
• Therapy is not usually needed and
2. Seen in African, Mediterranean, and prognosis is good
Middle Eastern populations; symptoms less
severe than sickle cell anemia but more
severe than Hgb C disease F.Hemoglobin D

3. No Hgb A is produced; approximately 50% a. Caused when glycine replaces glutamic


Hgb S and 50% Hgb C are produced. acid at position 121 on the beta chain
Compensatory Hgb F may be elevated up to
7%. b. Found more commonly in Middle Eastern
and Indian populations
4. Laboratory
✓ Moderate to severe c. Both homozygous and heterozygous
normocytic/normochromic anemia conditions are asymptomatic
with target cells; characterized by SC
crystals; may see rare sickle cells or C d. Hgb D migrates with Hgb S and Hgb G on
crystals alkaline hemoglobin electrophoresis.

✓ positive hemoglobin solubility Combination Disorders


screening test • Mutations may occur in combination
because of inheritance of different genetic
E.Hemoglobin E mutations from each parent

a. Caused when lysine replaces glutamic acid • Each compound disorder has variable
at position 26 on the beta chain symptoms and severities

b. Found more commonly in Southeast Asian, • Some examples include Hgb SC disease,
African, and African-American populations Hgb S–b thalassemia, Hgb C–Harlem

c. Homozygous condition results in mild


anemia with microcytes and target cells;
heterozygotes are asymptomatic.
XI. THALASSEMIAS

Introducton A) Beta-Thalassemia

- Disorders caused by genetic mutations that - Thalassemia caused by mutations or


lead to quantitative changes that lead into deletions in the b-globin genes
decrease rate of synthesis of a structurally
normal globin chain (quantitative defect) 1. Major/homozygous (Cooley anemia)
resulting in an imbalance of globin chain
synthesis a. Markedly decreased rate of synthesis or
absence of both beta chains results in an
✓ Types and severity of thalassemia excess of alpha chains; no Hgb A can be
depends on the globin gene mutated produced; compensate with up to 90% Hgb
(alpha or beta ) and the number of F.
genes affected by the mutation
b. Excess alpha chains precipitate on the RBC
✓ Alpha -Globin is coded for on membrane, form Heinz bodies, and cause
chromosome 16 , A normal genotype rigidity; destroyed in the bone marrow or
is designated as aa/aa removed by the spleen

✓ Beta -Globin is coded for on c. Symptomatic by 6 months of age;


chromosome 11 hepatosplenomegaly, stunted growth,
jaundice; prominent facial bones, especially
✓ Severity varies from no clinical the cheek and jaw; iron overload from RBC
abnormalities to transfusion- destruction and multiple transfusions cause
dependent to fatal organ failure

✓ Thalassemia major: Severe anemia; d. Laboratory:


either no alpha or no beta chains ✓ Severe microcytic/hypochromic
produced anemia
✓ target cells, teardrops, many nRBCs
✓ Thalassemia minor/trait: Mild ✓ basophilic stippling, Howell-Jolly
anemia; sufficient alpha and beta bodies, Pappenheimer bodies, Heinz
chains produced to make normal bodies
hemoglobins A, A2, and F, but may ✓ increased serum iron and increased
be in abnormal amounts bilirubin
2. HgbH disease
2. Minor/heterozygous
a. Three alpha genes are deleted. Decrease in
a. Decreased rate of synthesis of one of the alpha chains leads to beta chain excess.
beta chains; other beta chain normal
b. Hemoglobin H ($4), an unstable
b. Laboratory: hemoglobin, is produced. Heinz bodies form
✓ Mild microcytic/hypochromic anemia, and rigid RBCs are destroyed in the spleen.
with a normal or slightly elevated RBC
count ✓ Distinguishing characteristics include:
moderate microcytic/hypochromic
✓ target cells, basophilic stippling anemia; up to 30% Hgb H; the rest is
Hgb A.
c. Hgb A is slightly decreased, but Hgb A2 is
slightly increased to compensate
3. Minor/trait

C. Alpha-Thalassemia a. Two alpha genes are deleted. Patients are


- Thalassemias caused by mutations or usually asymptomatic and discovered
deletions in the a-globin genes accidentally.

-Four main groups ✓ Up to 6% Hgb Bart's in newborns


1. Major (hydrops fetalis) may be helpful in diagnosis; absent
by 3 months of age
a. All four alpha genes are deleted; no
normal hemoglobins are produced. b. Mild microcytic/hypochromic anemia
often with a high RBC count and target cells
b. 80% hemoglobin Bart's (74) produced;
cannot carry oxygen; incompatible with life;
die in utero or shortly after birth . 4. Silent carrier

✓ Not compatible with life as the blood a. One alpha gene is deleted. Patients are
is unable to oxygenate tissues asymptomatic and are often not diagnosed
because of high O2 affinity of Hgb unless gene analysis is done.
Bart
LEUKOCYTIC DISORDERS

I. NONNEOPLASTIC DISORDERS 3. Neutrophilic leukemoid reaction (NLR)


A) Nonmalignant Granulocytic Disorders
a. Blood picture mimics that seen in chronic
1. Shift/physiologic/pseudoneutrophilia myelogenous leukemia.

a. Redistribution of the blood pools causes a b. Benign, extreme response to a specific


short-term increase in the total WBC count agent or stimulus
and in the absolute number of neutrophils in
the circulating granulocyte pool. c There is a shift to the left with toxic changes
b. Caused by to the neutrophils.
✓ Exercise
✓ Stress 4. Neutropenia
✓ Pain a. Decrease in absolute number of
✓ Pregnancy neutrophils
b. risk of infection increases as neutropenia
c. The total blood granulocyte pool in the worsens
body has not changed. c. Congenital neutropenia may occur in
✓ The bone marrow has not released several disorders. The disorders are relatively
immature neutrophils. rare
✓ There are no toxic changes, and there d.Acquired neutropenia occurs more
is no shift to the left. commonly usually Due to bone marrow
production defects like:
2. Pathologic neutrophilia • Bone marrow injury (aplastic
anemia)
A) Neutrophils leave the circulating pool, • bone marrow infiltration
enter the marginating pool, and then move • DNA synthesis defects due to vitamin
to the tissues in response to tissue damage. B12 or folate deficiency
• Hypersplenism causes neutrophils to
Causes of Neutrophilia : be removed from circulation.
✓ Increase in relative and/or absolute Other reason :
numbers of cells (>8.7109/L or >70%) ✓ anti-neutrophil antibodies
✓ Infections, particularly bacteria ✓ chemotherapy and radiation
✓ Inflammation ✓ severe infections . Cell production is
✓ Medications unable to keep up with cell
✓ Increases may be caused by some consumption
neoplasms,particularly ✓ Intrinsic defects—Fanconi’s,
myeloproliferative disorders Kostmann’s, cyclic neutropenia,
Chédiak-Higashi
✓ Immune-mediated—collagen
vascular disorders, RA, AIDS
5.Eosinophilia 8. Basopenia

a. Increase in the absolute number of a. Decrease in the absolute number of


eosinophils basophils associated with inflammatory
states and following immunologic reactions
b.Caused by :
1) Parasitic infections, allergic reactions, b.Difficult to diagnose because of their
chronic inflammation normally low reference range

2) Chronic myelogenous leukemia, including B) Nonmalignant Monocytic Disorders


early maturation stages, Hodgkin disease,
tumors and chronic eosinophilic leukemia 1.Monocytosis

6.Eosinopenia ✓ Absolute counts are increased above


1.1109/L
a. Decrease in the absolute number of ✓ Often seen as patients are
eosinophils recovering from iacute bacterial
b. Seen in acute inflammation and infections and recovery following
inflammatory reactions that cause release of marrow suppression by drugs
glucocorticosteroids and epinephrine
✓ May be increased in some solid
7. Basophilia tumors and hematologic
malignancies, including acute
a. Increase in the absolute number of monocytic leukemia, acute
basophils myelomonocytic leukemia or chronic
myelomonocytic leukemia
b. caused by: ✓ Autoimmune disorders (systemic
1) Type I hypersensitivity reactions lupus erythematosus, rheumatoid
arthritis)
2) Chronic myelogenous leukemia, including
early maturation stages, polycythemia vera 2. Monocytopenia

3) Relative transient basophilia can be seen a. Decrease in the absolute number of


in patients on hematopoietic growth factors. monocytes
b. Associated with stem cell disorders such
as aplastic anemia

C. Nonmalignant Lymphocytosis
Associated with Viral Infections

2.Lymphocytopenia
1.Lymphocytosis
• Absolute counts are decreased
• Absolute counts are increased • Decreased counts are often seen in
• viral infections -hepatitis, influenza, immunodeficiencies, particularly human
mumps, measles, rubella, and varicella immunodeficiency virus infection and also
during steroid treatment
• A more normal lymphocyte morphology is
seen in disorders (non viral )such as
Bordetella pertussis infection ,brucellosis, I.Inherited Abnormalities of Neutrophils
toxoplasmosis a. May-Hegglin anomaly

Viral Infection Causing Lymphocytosis 1) Autosomal dominant inheritance


a. Infectious mononucleosis mutation of the MYH9 gene
✓ .Epstein-Barr virus (EBV) infects B
lymphocytes. 2) Large, crystalline, Dohle-like inclusions in
✓ Transmitted through nasopharyngeal the cytoplasm of neutrophils on Wright's
secretions stain; gray-blue and spindle (cigar) shaped
✓ Positive heterophile antibody test
✓ Lymphocytes usually >50% of the 3) Morphologically abnormal, but
WBCs, with 20% being reactive T functionally normal
lymphocytes attacking affected B
lymphocytes 4)Presence of thrombocytopenia and giant
b. Cytomegalovirus (CMV) platelets ,Usually asymptomatic, although
patients may exhibit bleeding as a result of
✓ Symptoms similar to infectious thrombocytopenia
mononucleosis
✓ Transmission is by blood transfusions b. Alder-Reilly anomaly
and saliva exchange 1) Autosomal recessive disorder leading
✓ 90% of lymphocytes can be reactive. to the inability to fully degrade
✓ Negative heterophile antibody test mucopolysaccharides
✓ Transfused blood products are often
tested for CMV.
2) Cells are filled with large, prominent e.Chronic granulomatous disease
granules composed of
mucopolysaccharides WBCs function 1. Mutations, either X-linked
normall recessive or autosomal recessive,
3) Must differentiate from toxic in the proteins coding for NADPH
granulation present in neutrophils only in oxidase
infectious conditions
2. Phagocytic cells produce
c. Pelger-Huet anomaly superoxide, which is needed for
the kill mechanism that targets
1. Autosomal dominant mutation of the many bacteria and fungi
lamin B receptor
2. Neutrophils are hyposegmented, with 3. Patients tend to have frequent
nuclei showing mature chromatin infections
3. Nuclei shapes are round/oval ,bands,or 4. Cells look normal but are unable
bilobed and separated by a thin filament to kill many bacteria or fungi,
4. Cells function normally, as granule leading to frequent infections
function is not impaired
5. Testing for the disorder uses the
d.Chediak-Higashi syndrome nitroblue tetrazolium reduction
test, in which patients with chronic
1. Autosomal recessive mutation of the granulomatous disease test
LYST gene that affects all cells with negative for the ability to reduce
lysosomal organelles the substance
2. All WBCs may show presence of large
lysosomal granules f. Leukocyte adhesion disorder (LAD)
3. Patients tend to die early in life as a
result of bacterial infections, because 1. Mutations in the genes needed to
cells do not function normally form cell adhesion molecules,
4. Patients also may exhibit bleeding, particularly b integrins
albinism, and neurologic issues
2. Three subtypes of LAD, all
affecting neutrophil adhesion

3. Patients have difficulties with


recurrent infections
III .Nuclear abnormalities of neutrophils

a. Hypersegmentation c.Dohle bodies


✓ Pale bluish inclusions in the
✓ characterized by 5 or more lobes in cytoplasm composed of rough
the neutrophil; associated with endoplasmic reticulum, usually
megaloblastic anemia due to vitamin associated with bacterial infections
B12 or folic acid deficiencies and inflammation

b. Hyposegmentation

✓ refers to a tendency in neutrophils to d.Vacuolization


have 1 or 2 lobes; may indicate an ✓ Vacuoles within the cytoplasm that
anomaly or a shift to the left. are often indicative of phagocytosis.
Examples : Vacuoles can be seen in bacterial or
fungal infections
1) Pelger-Huet anomaly ✓ may appear as artifact in old samples
✓ does not mature past the two-lobed
stage. e. Toxic granulation
✓ Must differentiate from a shift to the ✓ Large bluish-black granules appearing
left associated with an infection (toxic in the cytoplasm, usually present in
changes); infection requires inflammation
treatment but PelgerHuet anomaly
(no toxic changes) does not.

2) Pseudo Pelger-Huet
✓ Acquired abnormality associated
with myeloproliferative disorders and
myelodysplastic syndromes; can also
be drug induced

II. MALIGNANT LEUKOCYTE DISORDERS


A. Introdcution e. Immune dysfunction
-Hematopoietic neoplasms have been
categorized by a World Health Organization ✓ Hereditary and acquired defects in
classification according to cell of origin, the immune system
cytogenetic and molecular abnormalities,
immunophenotype, and clinical features.
3. Can be classified by stem cell involved and
1. A malignant clone of cells proliferate that length of clinical course
do not respond to normal regulatory
mechanisms. a. Lymphoproliferative disorders—acute or
a. Leukemia originates in the bone marrow chronic
and is initially systemic.
b. Lymphoma originates in lymphoid tissue b. Myeloproliferative disorders—acute or
and is initially localized. chronic

2. Etiology remains unclear. Multiple theories 4. Bone marrow examination used to aid in
exist about oncogene activation, which most diagnosis
likely includes multiple factors: ✓ Optimal sample for examination
includes both the aspirate and core
a. Viral biopsy specimen
✓ Viruses can suppress immune
function or activate oncogenes (HTLV- ✓ Posterior superior iliac crest most
I,II,V)andHIV-l. commonly used; less commonly used
anterior iliac crest or sternum
b. Bone marrow damage B. Comparison of Acute and Chronic
✓ Radiation, chemicals, and Leukemias
malignancies secondary to cancer
treatments 1. Duration
a. Acute
c. Chromosome defects ✓ Survival is weeks to months without
✓ Some chromosomal abnormalities are treatment; death is due to infection
diagnostic for leukemic subtypes; and bleeding.
t(15;17) is diagnostic for acute
promyelocytic leukemia. b. Chronic
✓ Survival is years without treatment.
d. Genetic factors
✓ Increased incidence in Down
syndrome, Fanconi, and others
2. Predominant cell type c. Both acute and chronic

a. Acute ✓ Unexplained weight loss or night


✓ Immature/blast cells predominate. sweats
✓ Splenomegaly, hepatomegaly,
1) AML has myeloblasts. lymphadenopathy
2) ALL has lymphoblasts.
4. Treatment
b. Chronic
✓ Maturing or mature cells predominate ✓ Chemotherapy used is dependent on
type of leukemia. Proper diagnosis is
1) CML has granulocytes. crucial.
2) CLL has lymphocytes. ✓ Radiation
✓ Bone marrow/stem cell transplant
3. Clinical manifestations and laboratory ✓ Supportive with transfusions of red
findings blood cells and platelets, antibiotics,
growth factors
a. Acute
✓ sudden onset; affects all ages C. French-American-British (FAB) and World
✓ Weakness and fatigue due to anemia Health Organization (WHO)
✓ Petechiae and bruising due to
thrombocytopenia There are 2 classification for Hematopoietic
✓ Fever and infection due to malignancy.
neutropenia
✓ Marrow blasts 1) FAB classification
− World Health Organization ✓ FAB defines acute leukemia as >30%
classification >20% bone marrow blasts.
− >30% based on French-American- ✓ FAB classification is easier to use and
British classification with cellularity is still widely taught.
>70% Based on:
A)cellular morphology
b. Chronic B) cytochemical stain results.
✓ frequently asymptomatic initially;
affects adults 2) WHO classification
✓ Marrow cellularity is >70%.
✓ WHO classification is now the
standard for diagnosis.
✓ WHO classification is now the
standard for diagnosis.

Based on : b. FABL2
✓ cellular morphology 1) Most common in adults
✓ cytochemical stains
✓ Cell markers 2) Large lymphoblasts, heterogeneous
✓ Cytogenetics appearance
✓ molecular abnormalities
✓ clinical syndrome c. FABL3 - Poor prognosis

I. ACUTE LYMPHOBLASTIC LEUKEMIA 1) Leukemic phase of Burkitt lymphoma


(ALL) 2) Seen in both adults and children

✓ Unregulated proliferation of the 3) Lymphoblasts are large and uniform with


lymphoid stem cell . prominent nucleoli; cytoplasm stains deeply
✓ Primarily affects children, but is also basophilic and may show vacuoles.
seen in older adults
4) ALL FAB L3s are of B cell lineage
Clinical symptoms:
✓ Fever, d. Burkitt lymphoma
✓ bone/joint pain,
✓ bleeding, 1) High-grade non-Hodgkin lymphoma phase
✓ Hepatosplenomegaly of FAB L3 leukemia
Laboratory:
✓ Neutropenia 2) Endemic in East Africa with high
✓ anemia, and thrombocytopenia association with Epstein-Barr virus; children
✓ variable WBC count, present with jaw/facial bone tumors
✓ hypercellular marrow with bone
marrow blasts >20% (WHO) or >30% Genetic translocations
(FAB) − are helpful in diagnosis. Common
✓ Lymphoblasts stain PAS positive ones include:
✓ Sudan black B and myeloperoxidase
negative a. FAB L3/Burkitt lymphoma—1(8;14) with a
rearrangement of the MYC oncogene
FAB classification of acute lymphoblastic b. Pre-B cell ALL associated with t(9;22); B
leukemia (ALL) cell ALL associated with
c. T cell ALL associated with t(7;ll)
a. FAB LI –best prognosis
1) Most common childhood leukemia (2- to
10-year peak); also found in young adults

II. Acute Myeloproliferative Disorders ✓ Accounts for 30% of the AMLs


✓ Increased urine/serum lysozyme
1. Platelets, erythrocytes, granulocytes, ✓ SBB, MPO, and specific and
and/or monocytes can be affected. nonspecific esterase positive
2. Found mainly in middle-aged adults;
also children < 1 year old b. FAB Ml (AML without maturation)

3. Clinical symptoms: ✓ shows 90% or more marrow


✓ Fever, malaise, weight loss, myeloblasts; may have Auer rods
petechiae, bruises, mild (fused primary granules)
hepatosplenomegaly c. FAB M2 (AML with maturation)

4. Laboratory: ✓ shows <90% marrow myeloblasts;


✓ Neutropenia, anemia, and may have Auer rods; chromosome
thrombocytopenia abnormality t(8;21)

✓ variable WBC count; hypercellular ✓ Both FAB Ml and FAB M2 are SBB,
marrow with bone marrow blasts MPO, and specific esterase positive.
>20% (WHO) or >30% (FAB)
B .Acute promyelocytic leukemia (APL; FAB
M3)
A). Acute myelogenous leukemia (AML) ✓ Characterized by >30% marrow
promyelocytes with bundles of Auer
a. FAB MO rods (faggot cells); heavy azurophilic
✓ stain negatively with the usual granulation
cytochemical stains, myeloperoxidase ✓ Accounts for 5% of the AMLs
(MPO), and Sudan black B (SBB). ✓ SBB, MPO, and specific esterase
Constitutes <5% of AMLs. positive
✓ Proliferation of unipotential stem cell ✓ Malignant normoblasts are PAS
CFU-GM that gives rise to both positive.
granulocytes and monocytes ✓ The myeloblasts are SBB and MPO
positive

C. Acute myelomonocytic leukemia (AMML;


FAB M4) F.Acute megakaryocytic leukemia (AMegL;
FAB M7)
✓ Characterized by >20% (WHO) or
>30% (FAB) marrow myeloblasts with ✓ Characterized by a proliferation of
>20% cells of monocytic origin; may megakaryoblasts and atypical
have Auer rods megakaryocytes in the bone marrow;
blasts may have cytoplasmic blebs

✓ Accounts for < 1 % of the AMLs


D.Acute monocytic leukemia (AMoL; FAB ✓ Marrow aspiration results in dry tap;
MS) blood shows pancytopenia
✓ Difficult to diagnose with
✓ Characterized by >20% (WHO) or cytochemical stains and positive for
>30% (FAB) marrow monoblasts plt marker
✓ Accounts for 10% of the AMLs
✓ Nonspecific esterase positive; CD 14 NOTE:
positive ✓ Bilineage leukemias: contain two cell
✓ Contains two variants: populations. One population
a) M5a is seen in children with >80% expresses myeloid antigens; the other
monoblasts in the bone marrow. population expresses lymphoid
antigens.
b) M5b is seen in middle-aged adults with
<80% monoblasts in the bone marrow. ✓ Biphenotypic leukemias occur when
myeloid and lymphoid antigens are
E. Acute erythroleukemia (AEL, Di Guglielmo expressed on the same cell; poor
syndrome; FAB M6) prognosis
✓ Characterized by >20% (WHO) or
>30% (FAB) marrow myeloblasts and
>50% dysplastic marrow
✓ normoblasts.
III. Chronic Myeloproliferative Disorders ✓ Philadelphia chromosome, t(9;22), is
present in virtually all patients. All cell
✓ Characterized by hypercellular lines are affected except
marrow, erythrocytosis, lymphocytes. The few who lack the
granulocytosis, and thrombocytosis chromosome have a worse prognosis.

✓ Defect of the myeloid stem cell


✓ Named for the cell line most greatly
affecteted B. Essential thrombocythemia (ET)
✓ All may terminate in acute leukemia.
✓ Characterized by proliferation of
A. Chronic myelogenous leukemia (CML) megakaryocytes
✓ Found mainly in adults 60 years of
✓ presents with proliferation of age and older
granulocytes. ✓ Laboratory: Platelets commonly
✓ Found mainly in adults 45 years of age greater than 1000 X 109/L, giant
and older; often diagnosed secondary forms, platelet function
to other conditions abnormalities, leukocytosis
Clinical symptoms:
✓ Weight loss, splenomegaly, fever, C. Polycythemia vera (PV)
night sweats, and malaise
✓ Bone marrow has an increased M:E ✓ Malignant hyperplasia of the
ratio. multipotential myeloid stem cell
causes increase in all cell lines
Laboratory: (polycythemia); erythrocytes most
✓ Blood findings include mild anemia greatly increased despite decreased
and WBC between 50 and 500 X erythropoietin (EPO); inappropriate
109/L,with all stages of granulocyte erythropoiesis
production (shift to the left), including
early forms of eosinophils and ✓ High blood viscosity can cause high
basophils blood pressure, stroke, and heart
✓ Myelocytes predominate; may have a attack.
few circulating blasts. ✓ Laboratory: Increased RBC (7-10 X
1012/L), hemoglobin (>20 g/dL), and
hematocrit (>60%) along with
✓ CML can mimic a neutrophilic increased leukocytes and platelets
leukemoid reaction (NLR). LAP score indicate polycythemia.
is used to differentiate; LAP is low in
CML and high in NLR.
✓ RBC mass is increased with a normal . Chronic lymphocytic leukemia (CLL)
plasma volume.
✓ Treatment is therapeutic phlebotomy, ✓ Found in adults over 60 years old;
splenectomy, and chemotherapy more common in males (2:1); survival
rate of 5-10 years
✓ PV is a chronic disease with a life
expectancy after diagnosis of up to 20 ✓ B cell malignancy (CD19, CD20
years positive)

E. Chronic idiopathic myelofibrosis ✓ Often asymptomatic and diagnosed


secondary to other conditions
✓ Myeloid stem cell disorder
characterized by proliferation of ✓ Laboratory:
erythroid, granulocytic, and Bone marrow hypercellular; blood shows
megakaryocytic precursors in absolute lymphocytosis of >5.0 X 109/L;
marrow with dyspoiesis homogeneous, small, hyperclumped
lymphocytes and smudge cells
✓ Progressive marrow fibrosis
✓ Laboratory: ✓ Anemia is not usually present unless
Anisocytosis, poikilocytosis with teardrop secondary to warm autoimmune
cells, leukoerythroblastic anemia hemolytic anemia (frequent
(immature neutrophils and nucleated complication).
RBCs in circulation); abnormal
morphology associated with all cell lines ✓ Bone marrow shows decreased M:E
ratio because of elevated lymphoid
IV. Chronic Lymphoproliferative Disorders cells.

✓ Neoplasms of mature lymphoid cells, NOTE: Small lymphocyte lymphoma (SLL) is


both B cells and T cells the lymphoma phase of CLL.

✓ Increases in mature lymphoid cells in


peripheral circulation (leukemic) or
in masses in the tissues (lymphoma)
2. Hairy cell leukemia (HCL) V. Lymphoid Malignancies

✓ Found in adults over 50 years old; 1. Plasma cell neoplasms


more common in males (7:1) ✓ B cell disorders leading to increases
in plasma cells
✓ B cell malignancy (CD19, CD20 ✓ Typically affects older adults
positive)
✓ Bone marrow Often exhibits areas of a. Multiple myeloma
fibrosis, leading to a dry tap on
aspiration. Hairy cells may be visible ✓ Monoclonal gammopathy causes B
on core biopsy cell production of excessive IgG
(most common) or IgA, with
✓ Clinical presentation decreased production of the other
Patients often present with massive immunoglobulins.
splenomegaly
✓ Identified on serum protein
electrophoresis by an "M"-spike in
✓ Laboratory: the gamma-globulin region;
Pancytopenia; cytoplasm of lymphocytes immunoglobulin class determined
shows hair-like projections; hairy cells are using immunoelectrophoresis
tartrate-resistant acid phosphatase (TRAP)
stain positive ✓ Excessive IgG or IgA production by
myeloma cells causes increased
3. Prolymphocytic leukemia (PLL) blood viscosity
✓ Laboratory:
✓ Found in adults; more common in Bone marrow plasma cells >30%
males marked rouleaux, increased erythrocyte
✓ Can be either B cell (most common) sedimentation rate (ESR)
or T cell malignancy
✓ Marked splenomegaly ✓ Bence Jones proteins (free light
✓ Laboratory: chains—kappa or lambda) found in
Characterized by lymphocytosis (>100 X the urine; toxic to tubular epithelial
109/L) with many prolymphocytes; cells; cause kidney damage
anemia and thrombocytopenia

✓ Both B and T cell types are aggressive


and respond poorly to treatment.
b. Waldenstrom macroglobulinemia A) Hodgkin lymphoma (classical)

✓ Monoclonal gammopathy causes B -Hodgkin lymphoma is a neoplasm of


cell production of excessive IgM defective B cells. Much of its pathology is
(macroglobulin) and decreased due to an associated inflammatory milieu
production of the other
immunoglobulins. ✓ 40% of lymphomas; seen in patients
between 15 and 35 years of age seen
✓ Found in adults over 60 years old more frequently in males; certain
subtypes have an Epstein-Barr virus
✓ Identified on serum protein (EBV) association
electrophoresis by an "M"-spike in the
gamma-globulin region; ✓ Reed-Sternberg (RS) cells found in
immunoglobulin class determined lymph node biopsy are large,
using immunoelectrophoresis multinucleated cells each with
prominent, large nucleoli; B cell
✓ Excessive IgM production causes lineage
increased blood viscosity ✓ Laboratory:
Mild anemia, eosinophilia, and
✓ Laboratory: monocytosis; increased LAP score and
Marked rouleaux, increased ESR, blue ESR during active disease
background to blood smear; plasmacytoid
lymphocytes, plasma cells, and ✓ Hodgkin lymphoma subtypes using
lymphocytes on blood smear WHO classification:

2. Lymphoma a) Nodular sclerosis—70% are this subtype;


lowest EBV association
✓ Proliferation of malignant cells in solid
lymphatic tissue b) Mixed cellularity—20% are this subtype;
✓ Initially localized; may spread to bone highest EBV association
marrow and blood
✓ Clinical symptom: Lymphadenopathy B) Non-Hodgkin lymphoma
NOTE: - Non-Hodgkin lymphomas most often arise
World Health Organization (WHO) groups from mature B cells. They are pathologically
the lymphomas into and clinically heterogeneous.
✓ Hodgkin, B cell T cell lymphomas are less common, also
✓ T/NK cell (non-Hodgkin) neoplasms heterogeneous, and often difficult to treat
successfully.
✓ WHO separates B cell and T/NK cell VI. Myelodysplastic Syndromes (MDSs)
neoplasms into conditions with
precursor cells or mature cells. ✓ -myelodysplastic syndromes are
heterogeneous disorders of
✓ Enlarged lymph nodes or differentiated myeloid cells. They
gastrointestinal (GI) tumors often are initially indolent but
✓ ultimately progress.
✓ B cell neoplasms are more common;
include Burkitt (lymphoma phase of ✓ Group of acquired clonal disorders
Burkitt leukemia), mantle cell, affecting the pluripotential stem cell;
follicular, and other lymphomas characterized by progressive blood
✓ Can be slow growing or very cytopenias despite bone marrow
aggressive hyperplasia

1.Mycosis fungoides (cutaneous T cell ✓ Dyspoiesis affects erythroid, myeloid,


lymphoma) and megakaryocytic cell lines.

✓ Classified by WHO as a T/NK cell ✓ MDS development can be triggered


neoplasm (non-Hodgkin lymphoma by chemotherapy, radiation, and
chemicals.
✓ Cutaneous lymphoma causes skin
itching, leading to ulcerative tumors. There are 5 subgroups of MDS using the FAB
classification scheme; up to 30% blasts in the
✓ Sezary syndrome, a variant of bone marrow
mycosis fungoides, presents as a
disseminated disease with 1.Refractory anemia (RA)
widespread skin involvement and ✓ Anemia that is refractory (not
circulating lymphoma cells. responsive) to therapy
✓ Laboratory:
✓ CD2, CDS, and CD4 positive Oval macrocytes, reticulocytopenia,
dyserythropoiesis; bone marrow blasts <5%
and peripheral blood blasts <1%
2.Refractory anemia with ringed 5. Refractory anemia with excess blasts in
sideroblasts (RARS) transformation (RAEB-t)

✓ This is the primary/idiopathic ✓ Laboratory:


sideroblastic anemia discussed with bone marrow blasts >20% but less than
the anemias. 30%; peripheral blood blasts >5%

✓ Ringed sideroblasts comprise more ✓ WHO classification reassigns RAEB-t


than 15% of bone marrow nucleated as an acute leukemia instead of a
cells. Signs of dyserythropoiesis, myelodysplastic syndrome because
neutropenia of the bone marrow blast percent.

✓ Laboratory: Similar to RA;


dimorphic erythrocytes. Note:
✓ WHO classification of MDS has
3 Chronic myelomonocytic leukemia additional groups (e.g., refractory
(CMML) cytopenia with multilineage
dysplasia, 5q deletion syndrome).
✓ The one MDS that usually presents
with leukocytosis ✓ WHO created the new category of
myelodysplastic/myeloproliferative
✓ Laboratory: Bone marrow blasts 5- disease, which includes the FAB's
20% and peripheral blood blasts <5% CMML.

4.Refractory anemia with excess blasts


(RAEB)
✓ Trilineage cytopenias common

✓ Laboratory: Bone marrow and


peripheral blood blasts are the same
as with CMML, but there is no
absolute monocytosis.

✓ The higher the blast percent, the


worse the prognosis.
Cytochemical Stains—Used in Diagnosis of Hematologic Disorders
1.Myeloperoxidase (MPO) ✓ Esterase enzymes found in
neutrophils monocytic cells negative
✓ Cells of the granulocytic series and to for this stain
a lesser degree the monocytic series b. Nonspecific esterase stains (alpha-
contain the enzyme peroxidase in naphthyl acetate and alpha-naphthyl
their granules that is detected by this butyrate)
stain. Auer rods also stain positive ✓ Esterase enzymes found in monocytes
,detects esterase enzyme present in
✓ lymphocytic cells are negative for monocytic cells
this stain
✓ Enzyme found in the primary granules ✓ granulocytic cells negative for these
of granulocytic cells stains
✓ Diagnostic Utility
- Differentiating AML versus ALL ✓ Diagnostic Utility
AML of myeloid versus monocytic origin
2. Sudan black B
Note:
✓ Stains phospholipids and The esterase stains may be useful in
lipoproteins found in primary and distinguishing acute leukemias that are of
secondary granules and in lysosomal myeloid origin (FAB Ml, M2, M3, M4) from
granules of monocytes those leukemias that are primarily cells of
monocytic origin (FAB M5).
✓ Diagnostic Utility
- Differentiating AML versus ALL 4. Periodic acid-Schiff (PAS)
✓ Used to stain Glycogen, mucoproteins
✓ Granulocytic cells and Auer rods stain and high-molecular weight
positive (blue-black granulation) carbohydrates
✓ lymphocytic cells are negative for
Sudan black B (reaction parallels ✓ PAS stains intracellular glycogen
MPO) bright pink.

3. Esterases - ✓ Immature lymphoid cells, malignant


There are 2 types erythroblasts, and megakaryocytic
A)Specific esterase stain (naphthol AS-D cells stain positive with this stain
chloroacetate esterase stain)
✓ myeloblasts and normal erythrocytic
✓ Detects esterase enzyme present in cells are negative with this stain
primary granules of granulocytic cells; ✓ Useful in diagnosis of erythroleukemia
(FAB M6) and acute lymphoblastic
leukemia
4. Leukocyte alkaline phosphatase ✓ Almost all blood cells contain the acid
(LAP) phosphatase enzyme and show
✓ Detects alkaline phosphatase enzyme positivity with acid phosphatase stain.
activity in primary granules of Once tartrate is added, staining is
neutrophils inhibited in most cells

✓ Differentiation of CML (low activity) ✓ Only hairy cells from hairy cell
and neutrophilic leukemoid reaction leukemia are resistant to inhibition
(high activity) with tartrate and continue to stain
positive; all other cells stain negative.
✓ LAP score
1) 100 neutrophils are graded on a scale ✓ Positive diagnosis of hairy cell
from 0 to 4+ based on stain intensity and leukemia
size of granules. Results are added
together.

2) Reference range is 13-130. 6. Perl's Prussian blue stain

✓ Clinical significance ✓ Free iron precipitates into small


1) Decreased LAP score: blue/green granules in mature
✓ CML, paryoxysmal nocturnal erythrocytes; cells are called
hemoglobinuria siderocytes.
2) Normal LAP score:
✓ Iron inclusions are called siderotic
✓ CML in remission or with infection,
Hodgkin lymphoma in remission, granules or Pappenheimer bodies
secondary polycythemia when visible with Wright's stain.

3) Increased LAP score: 7.Terminal deoxynucleotidyl transferase


✓ Neutrophilic leukemoid reaction, (TdT)
polycythemia vera, CML in blast crisis,
late trimester pregnancy ✓ Stains DNA polymerase in cell nuclei
✓ Identification of lymphoblasts in ALL
5. Tartrate-resistant acid phosphatase stain ✓ Positive reaction in Lymphoblasts
(TRAP) ✓ Negative reaction Myeloblasts and
✓ Isoenzymes of acid phosphatase monoblasts
Part II.

Hemostasis and Thrombosis


Hemostasis respond
✓ Injury to endothelium (or vessel)
✓ Primary hemostasis (formation of primary hemostatic plug, platelets have the main role)
Secondary hemostasis (formation of fibrin clot, coagulation proteins are the major
contributor)
✓ Fibrinolysis (removal of clot)

ROLE OF VASCULATURE
• Hemostasis usually occurs in thearterioles andvenules
✓ Endothelial cells line lumen
✓ Luminal side coated by glycocalyx (carbohydrates and proteins)
✓ Abluminal side is attached to basement membrane (type IV collagen and proteins)

• Vessels are nonthrombotic under normal circumstances


• Damaged vessels are prothrombotic
✓ Exposure of subendothelium: Collagen—platelet activation
✓ Secretion of platelet activating factor
✓ Secretion of von Willebrand factor (vWF): Platelet adhesion
✓ Release of tissue factor: Aids in secondary hemostasis activation

ROLE OF PLATELETS

• Characteristics
✓ Circulate as inert cell fragments
✓ Repel each other and endothelial lining (nonthrombotic property)
✓ Become activated after an injury
✓ After activation, platelets interact with other platelets and the damaged vessel wall
Platelet Characteristics
✓ The reference range for healthy individuals is 150-450 X 109/L or approximately 7-21
per high power field. Two-thirds of available platelets are in circulation; one-third is
stored in the spleen.
✓ Life span of 8-12 days; shorter in certain disease states
✓ With Wright's stain, platelets stain gray-blue with purple granules.
✓ Platelets are found in the bone marrow, spleen, and blood vessels; in the blood vessels
platelets function in hemostasis.
✓ Originate from the same progenitor cell as the erythroid and myeloid series
✓ Giant platelets indicate premature release from the bone marrow and result from
increased demand.
✓ Immature platelets are found in the peripheral blood in certain diseases like acute
megakaryocytic leukemia, myelodysplastic syndrome .

Platelet Ultrastructure

The platelet is divided into arbitrary zones described by location and function
Major Zones Location
✓ Peripheral zone
✓ its located in Glycocalyx Cytoplasmic membrane Open canalicular system Sub membranous
area

Major fuction :

✓ Factor V: Component of prothrombinase complex, attachment site for factor X on platelet


surface
✓ vWF: Transports factor VIII, mediates adhesion between platelets via GPIb/IX
✓ Fibrinogen: Converted to fibrin in final clot formation stages
✓ GPIb/IX: Platelet receptor for vWF
✓ GPIIb/IIIa: Platelet receptor for fibrinogen

2.Structural zone
✓ Is found Circumferential and throughout the platelet

Major function :

✓ Microtubules
✓ Microfilament
✓ Intermediate filaments
✓ All involved in maintenance of shape and shape change on platelet activation

3. Organelle zone

Internally located

Major components .

✓ Granules a (50-80 per platelet):


✓ Dense (3-8 per platelet)
✓ Lysosomal granules: Hydrolytic Peroxisomes Mitochondria Glycogen particles

4. Membrane systems
Location
✓ Surface connected open canalicular system (SCCS, OCS)
✓ Dense tubular system (DTS)
Fuction

✓ SCCS: Interior of platelet and connects to platelet surface; allows substances to enter
platelet and others to exit; important in storage and secretion; serves as source of
surface membrane after activation
✓ DTS: Does not connect to platelet surface, primarily a source of ionized calcium, site of
prostaglandin and thromboxane synthesis
PRIMARY HEMOSTASIS
A)Platelet Adhesion
✓ Major interaction is the binding of platelet receptor glycoprotein Ib (GPIb)/IX to vWF,
which binds to collagen
✓ vWF: Stored in a-granules in platelets and WeibelPalade bodies in endothelial cells
✓ Important step that triggers several events leading to platelet activation
B) Platelet Activation

✓ Triggered after platelet adhesion or exposure to agonist


✓ Results: Shape change, altered orientation of phospholipids, new receptor expression,
changes in biochemistry
✓ Platelet agonists: Collagen, adenosine diphosphate (ADP), thrombin, epinephrine,
thromboxane A2 (TXA2), arachidonic acid
✓ TXA2: Synthesized from arachidonic acid by cyclooxygenase and thromboxane synthase,
stimulates platelet granule secretion, enhances vasoconstriction; if blocked, secretion is
impaired.
✓ Aspirin blocks cyclooxygenase

C) Platelet Aggregation
✓ vWF binding to GPIb/IX activates an intracellular signaling pathway that results in the
activation of GPIIb/ IIIa, which then binds to fibrinogen
✓ vWF binding GPIb/IX #Intracellular signaling GPIIb/IIIa activation and binding to
fibrinogen
✓ Fibrinogen forms bridges to other GPIIb/IIIa receptors on other activated platelets
,resulting in platelet aggregates; Ca2+ is needed for aggregation
✓ Fibrinogen and Ca2+ are delivered locally from granules and dense tubular system

There are 2 thypes of aggregation in vitro :


✓ Primary: Loose aggregation, reversible if stimulus is not sufficient
✓ Secondary: Irreversible provided sufficient stimulus; occurs after internal ADP release,
TXA2 synthesis and release, further stimulation then occurs
SECONDARY HEMOSTASIS
✓ Through a series of enzymatic reactions, the primary platelet plug is reinforced by fibrin
✓ Secondary hemostasis is a complex system of procoagulant activities and control
activities to contain and limit clot formation
✓ Vitamin K–dependent factors:II,VII,IX,X,proteinC, and protein S
✓ Vitamin K–dependent factors are not functional unless an additional carboxyl group
(COOH) is added to the g-carbon of the glutamic acid residues. This reaction is called g-
carboxylation and is dependent on vitamin K.
The coagulation cascade has two pathways
✓ intrinsic
✓ Extrinsic

Note :They both shares a common final pathway , The end-point of the common pathway is the
formation of a fibrin clot that reinforces the platelet plug

A)Intrinsic Pathway
✓ Activation of contact factors when they come in to contact with negatively charged
surfaces like Glass, kaolin, ellagic acid
✓ Not dependent on calcium
✓ Deficiency of contact factors (XII, PK, and HK) does not lead to in vivo bleeding issues.
Deficiency of XI is associated with bleeding abnormalities in approximately 50% of
individuals
✓ Contact factors are involved in activation of fibrinolysis, complement activation, kinin
formation, inflammation, and angiogenesis

B)Extrinsic Pathway
✓ Damage to the vessel results in the exposure of tissue factor on the surface of
nonvascular cells
✓ VIIa ndVIIa bind to tissue factor in the presence ofcalcium to form the VIIa/tissue factor
complex, also called extrinsic Xase, and the extrinsic pathway is thus activated.
✓ Extrinsic Xase also can activate IX in the intrinsic pathway
C) common pathway
✓ Begins with factor X activation by either the extrinsic (main in vivo) or intrinsic pathway.
It includes factors X (Stuart-Prower), V (proaccelerin/labile factor), II (prothrombin), and
I (fibrinogen).
✓ End result: Formation of fibrin clot

THE FIBRINOLYTIC SYSTEM

✓ Process of breaking down fibrin clot through hydrolysis of fibrin


✓ Activation of system occurs when the intrinsic pathway is activated
✓ Fibrinolytic System: Keeps blood vessels clear and is important in clot dissolution. During this
process, plasminogen is activated to plasmin.

✓ Plasminogen

1. Glycoprotein produced in the liver

2. Zymogen (inert) found in the plasma

3. Converted to plasmin by plasminogen activators

a. Intrinsic activators are Xlla, kallikrein, and HMWK.

b. Extrinsic activators are tissue-type plasminogen activator (t-PA) and urokinase-type plasminogen
activator (u-PA)

c. Exogenous activators (therapeutic agents) include t-PA, streptokinase, and urokinase. They are
administered to lyse existing clots

✓ Plasmin

1. Not normally found in circulation; the precursor plasminogen is found in circulation

2. Degrades fibrin clots (fibrinolysis), fibrinogen (fibrinogenolysis), factors V and VIII

3. Activates the complement system

REGULATORY PROTEINS OF COAGULATION AND FIBRINOLYSIS


A. Antithrombin (AT)
1. Produced in the liver
2. Principal inhibitor of coagulation
3. Inhibits the serine proteases

4. Therapeutic heparin enhances the action of antithrombin.

B. Proteins C and S
1. Vitamin K-dependent regulatory proteins

2. Activated when thrombin binds to thrombomodulin on the endothelial cell surface


3. Inhibit factors V and VIII to provide negative feedback on the cascade

C. Tissue Factor Pathway Inhibitor

✓ Inhibits factor Vila-tissue factor complex


D. alpha 2-Macroglobulin:
✓ inhibits thrombin, Xa, kallikrein, and plasmin
E. alpha 1-Antitrypsin:
✓ Inhibits XIa and inactivates plasmin

F. C1 Inhibitor
✓ Inhibits Cl from the complement cascade, and Xlla, XIa, kallikrein, and plasmin
G. alpha 2-Antiplasmin
✓ Principal inhibitor of fibrinolysis; neutralizes plasmin
H. PAI-1 (plasminogen activator inhibitor-1)
1. Important inhibitor of fibrinolysis
2. Prevents activation of plasminogen by t-PA; released from endothelial cells upon damage

Laboratory Analysis of Platelets


1. Quantitative a. Platelet numbers
✓ Automated instrumentation,
✓ hemacytometer counts,
✓ blood smear estimates

2. Qualitative
✓ Bleeding time will detect defects in adhesion, release, and aggregation.
✓ Platelet aggregation studies detect platelet function abnormalities. Aggregating agents
used include ADP, epinephrine, collagen, thrombin, and ristocetin.
✓ vWF:Ag (antigenic) and vWF:RCo (activity) assays are used to assess von Willebrand
factor

DISEASES AND CONDITIONS ASSOCIATED WITH THROMBOCYTES


Hereditary Adhesion Defects
1 . von Willebrand disease
✓ Most common hereditary hemorrhagic disorder; autosomal-dominant inheritance
✓ Lacks von Willebrand factor, which is needed for platelets to adhere to collagen in
damaged vessels and is a carrier protein for coagulation factor VIILC
✓ Decreased platelet adhesion causes mucous membrane bleeding that is variable in
severity.
Laboratory:

✓ Normal platelet count


✓ prolonged bleeding time
✓ decreased aggregation response to ristocetin
✓ variable aPTT, normal PT, decreased vWF:RCo, vWF:Ag, and VIII:C

2. Bernard-Soulier syndrome
Giant platelets (increased MPV) that lack glycoprotein Ib receptor; adhesion defect due to
faulty binding of the platelet to von Willebrand factor
Laboratory:
✓ Variable platelet count
✓ platelet anisocytosis (increased PDW)
✓ prolonged bleeding time
✓ decreased aggregation response to ristocetin, normal aPTT and PT

Hereditary Aggregation and Clot Retraction Defect

1 . Glanzmann thrombasthenia
✓ Hemorrhagic disorder seen in populations where consanguinity is prevalent
✓ Lack of glycoprotein Hb/IIIa, the fibrinogen binding receptor
✓ Inability of fibrinogen to bind with platelets causes aggregation defect; lack of
thrombasthenin/actomyosin causes clot retraction defect.

Laboratory: Decreased aggregation response with ADP, epinephrine, and collagen, normal
response with ristocetin

Storage Pool Defects: Deficiency of One or More Types of Storage Granules


1 . Gray-platelet syndrome
✓ is characterized by large platelets, thrombocytopenia, and an absence of alpha granules.
✓ Patients are prone to lifelong mild bleeding tendencies.
2. Wiskott-Aldrich syndrome
✓ is characterized by small platelets (low MPV), thrombocytopenia, and a decreased
amount of alpha granules and dense bodies.
✓ Patients are prone to hemorrhage and recurrent infections.
3. Hermansky-Pudlak
✓ syndrome is characterized by a lack of dense body granules.
✓ Patients exhibit oculocutaneous albinism and are prone to hemorrhage

Acquired Defects
1. Drugs
a. Aspirin and nonsteroidal anti-inflammatory drugs

✓ interfere with the cyclooxygenase enzymes, preventing thromboxane A2 synthesis and


subsequent aggregation.

b. Clopidogrel bisulfate (Plavix®) and ticlopidine

✓ are adenosine diphosphate (ADP) receptor inhibitors. The blockage of this receptor
inhibits platelet aggregation
c. Eptifibatide and similar antiplatelet medications
✓ block Ilb/IIIa glycoprotein receptors, preventing aggregaties

2. Myeloproliferative disorders and uremia are examples of diseases that can cause platelet
dysfunction.

Quantitative Platelet Disorders


1. Primary thrombocytosis

✓ Uncontrolled, malignant proliferation of platelets, not in response to thrombopoietin


✓ can be caused by essential thrombocythemia, polycythemia vera, and chronic
myelocytic leukemia
✓ Platelet counts can be >1000 X 109/L.
✓ Associated with either hemorrhagic or thrombotic complications
2. Secondary (reactive) thrombocytosis
It is characterized by increased platelet production, usually in response to thrombopoietin.
Platelet count is elevated, but usually <1000 X 109/L. Can result from:

a. Chronic and acute inflammatory disease (e.g., tuberculosis, cirrhosis)


b. Iron deficiency: Iron regulates thrombopoiesis by inhibiting thrombopoietin; deficiency
causes increased thrombopoietin and stimulates thrombopoiesis
c. Rapid blood regeneration due to hemolytic anemia and acute blood loss
d. Exercise, prematurity, and response to drugs

3. Thrombocytopenia
Decrease in the number of platelets, which can result from the following:

a. Megakaryocyte hypoproliferation: Caused by chemotherapy, marrow replacement by


malignant cells, aplastic anemia, drug and alcohol abuse
b. Ineffective thrombopoiesis: Caused by megaloblastic anemias
c. Increased loss/destruction - due to immune or non immune causes

4) Thrombocytosis
✓ Primary thrombocytosis: Uncontrolled/autonomous proliferation of megakaryocytes,
hemorrhagic or thrombotic episodes, platelet count greater than 1000 x 10*9
✓ Reactive (secondary) thrombocytosis: Result of another condition ,variable causes and
platelet counts

5.Falsely low platelet counts


Platelet satellitosis:
✓ Platelets can adhere to neutrophils when exposed to EDTA. Redraw in sodium citrate to
correct; multiply obtained platelet count by 1.1 to correct for dilution factor in sodium
citrate tube.
EDTA-dependent platelet agglutinins:
✓ Platelets can adhere to each other when exposed to EDTA. Correction of the problem is
the same as for platelet satellitosis.

THROMBOTIC DISORDERS
A. Primary Thrombotic Disorders
1. Deficiency in regulatory proteins
a. Antithrombin (AT) deficiency

✓ Genetic deficiency occurs about 1:2000 in the general population; associated with deep
vein thrombosis and pulmonary embolism
✓ Serine proteases not inhibited; negative feedback to cascade impaired
Laboratory:

✓ Antithrombin activity assay (antigenic testing less common)

b. Protein C or Protein S deficiencies


✓ Vitamin K-dependent regulatory proteins that inactivate factors V and VIII
✓ Can cause superficial and deep vein thrombosis and/or pulmonary embolism

Laboratory:
✓ Immunologic and functional testing to diagnose

2.Genetic mutations
a. Factor V Leiden (Activated Protein C Resistance—APCR)
✓ Most common hereditary cause of thrombosis; caused by an amino acid substitution
✓ Protein C is incapable of inactivating factor V Leiden, causing thrombin generation and
subsequent fibrin clot formation.
Laboratory:
✓ PCR-based molecular assay to single-point mutation in the gene for factor V

b. Prothrombin gene mutation 20210


✓ Second most common hereditary cause of thrombosis; caused by an amino acid
substitution
✓ May have slightly elevated prothrombin level

Laboratory:
✓ PCR-based molecular assay

c. Dysfibrinogenemia

✓ Autosomal-dominant trait; abnormal structure of fibrinogen; caused by gene mutations


✓ Associated with either bleeding or thrombosis; dependent on the specific gene
mutation

B. Secondary Thrombotic Disorders


1. Lupus anticoagulant and anticardiolipin antibodies
✓ The body develops autoantibodies against platelet phospholipids; etiology is unknown.

2. Post-operative status:
✓ Thrombotic event starts after tissue factor release during surgery, activating the
extrinsic coagulation (dominant in vivo) pathway
3. Malignancy
✓ Risk of malignancy increases because of the release of thromboplastic substances by
neoplastic cells.
4.Pregnancy

✓ The placenta is rich in tissue factor, which may enhance thrombosis during pregnancy,
especially high-risk patients having caesarian section delivery.
✓ Factor V and VIII levels increase, contributing to clot formation.

5. Estrogen/oral contraceptives:
✓ Increase risk of venous thrombosis and renal artery thrombosis
6. Morbid obesity
✓ Results in decreased AT levels and increased PAI-1, causing thrombosis

7. Hyperhomocysteinemia:
✓ This disorder is linked to atherosclerosis, resulting in arterial and venous
thromboembolism.
✓ Mechanisms are not fully understood but may be associated with a reduction in the
localized activation of the protein C pathway.

Differential Diagnosis of Abnormal Coagulation Screening Tests


Assessment of Primary Hemostasis
1. Bleeding time(BT)
✓ A small incision is made in the arm ,and the time needed for the platelets to stop
bleeding is measured in minutes
✓ Test is rarely performed because it is affected by many factors like depth of incision
,platelet count, pressure on arm.

2.Plateletaggregation
✓ The ability of platelets to aggregate in the presence of specific agonists (ADP
,arachidonicacid ,collagen ,epinephrine, ristocetin) is measured using a photo optical
instrument
✓ Patients must refrain from aspirin containing products for 7-10days before testing.
✓ Specimenmustbetestedwithin4hrof collection
✓ Aggregation is measured by a decrease in optical dens

3. Platelet function analyzer (PFA)


✓ Platelet function is assessed in whole blood by passing blood through cartridge
containing agonists(collagen/ epinephrine and collagen/ADP)
✓ The time needed for the platelets to occlude the aperture is measured
✓ A longer closure time in the presence of specific agonists may indicate a platelet
disorder
Assessment of Secondary Hemostasis
1)Abnormal Activated Partial Thromboplastin Time (APTT)
✓ Alone Associated with bleeding: VIII, IX, and XI defects
✓ Not associated with bleeding: XII, prekallikrein (PK), high molecular weight kininogen,
lupus anticoagulants Abnormal
2)Prothrombin Time (PT)
✓ Alone Factor VII defects

3)Combined Abnormal APTT and PT


✓ Medical conditions: Anticoagulants, disseminated intravascular coagulation (DIC), liver
disease, vitamin K deficiency, massive transfusion Rarely dysfibrinogenemia; factor X, V,
and II defects

HEMORRHAGIC DISORDERS
Hemorrhagic disorder can be inherited or acquired
✓ Inherited Disorders: Generally affect only one hemostatic component (e.g., factor VIII)
✓ Acquired Disorders: Involve multiple hemostatic components or pathways (e.g.,
warfarin therapy, liver disease)
✓ Hemorrhagic Symptoms: Associated with defects in secondary hemostasis; include
bleeding into deep tissues, joints, abdominal and other body cavities

Inherited Intrinsic Pathway Hemorrhagic Disorders


1. von Willebrand disease
✓ Autosomal-dominant trait
✓ Most common hereditary bleeding disorder; abnormalities in both primary and
secondary hemostasis
✓ Caused by a defect in von Willebrand factor that is needed for platelet adhesion to
collagen in primary hemostasis. vWF is also the carrier protein for factor VIII:C in
secondary hemostasis.
Clinical:

✓ Mild to moderate bleeding dependent of vWF and VIILC levels; menorrhagia common
symptom in women
Laboratory:
✓ Decreased vWF:RCo, vWF:Ag, and VIII:C; abnormal platelet aggregation with ristocetin,
variable aPTT (often prolonged because of decreased VIILC), and prolonged bleeding
time
Treatment:
✓ Factor VIII concentrates; DDAVP (deamino-D-argininevasopressin) used to raise plasma
levels of vWF and VIILC

2. Factor VIII :C (hemophilia A, classic hemophilia) deficiency


✓ Sex-linked disorder transmitted on the X chromosome by carrier women to their son
✓ Accounts for 80% of the hemophilias; second most common hereditary bleeding
disorder
✓ Many new cases of hemophilia A result from spontaneous mutations.
Clinical:
✓ Bleeding symptoms are proportional to the degree of the factor deficiency.
✓ Spontaneous bleeding occurs often and is especially bad in joint regions (hemarthrosis).
Laboratory:
✓ Prolonged aPTT only, factor VIII:C assay to confirm
Treatment:

✓ Cryoprecipitate and factor VIII concentrates are used; in mild cases


✓ DDAVP can be used to stimulate the release of VIII:C and vWF from stored reserves
Note: About 15-20% of patients will develop a factor VIII inhibitor; it is associated with a
bleeding tendency and worse prognosis.

3.Factor IX (hemophilia B, Christmas disease) deficiency


✓ Sex-linked recessive trait
✓ Accounts for 20% of the hemophilias; third most common hereditary bleeding disorder
Clinical

✓ Bleeding symptoms are similar to those seen in hemophilia A.


Laboratory:
✓ Prolonged aPTT only; factor IX assay to confirm
Treatment:

✓ Fresh frozen plasma (FFP) or factor IX concentrates

4. Factor XI (hemophilia C) deficiency


✓ Mainly seen in the Ashkenazi Jewish population
✓ Characterized by clinical bleeding that is asymptomatic until surgery or trauma
Laboratory:
✓ Prolonged aPTT only; factor XI assay to confirm

Inherited Extrinsic and Common Pathway Hemorrhagic Disorders

1. Factor VII (stable factor) deficiency


✓ Autosomal-recessive trait
✓ Clinical soft tissue bleeding
✓ Laboratory: Prolonged PT only

2. Factor X (Stuart-Prower) deficiency


✓ Autosomal-recessive trait
✓ Clinical: Soft tissue bleeding and chronic bruising
✓ Laboratory: Prolonged PT and aPTT

3. Factor V (Owren disease, labile factor) deficiency


✓ Autosomal-recessive trait
✓ Clinical: Mild to moderate bleeding symptoms
✓ Laboratory: Prolonged PT and aPTT
4.Factor II (prothrombin) deficiency
✓ Autosomal-recessive trait
✓ Clinical: Mild bleeding symptoms
✓ Laboratory: Prolonged PT and aPTT

5. Factor I (fibrinogen) deficiency


a. Autosomal-recessive trait; results from the following inherited disorders:
1) Afibrinogenemia: Inherited lack of fibrinogen; severe bleeding symptoms
2) Hypofibrinogenemia: Inherited deficiency of fibrinogen; bleeding symptoms correlate with
fibrinogen concentration
Clinical:
✓ Spontaneous bleeding of mucosa, intestines, and intracranial sites
Laboratory:
✓ Prolonged bleeding time (fibrinogen bridges do not form; platelet aggregation defect),
decreased fibrinogen concentration, and prolonged PT, aPTT, and thrombin time

6. Factor XIII (fibrin-stabilizing factor) deficiency


Autosomal-recessive trait

Clinical
✓ Spontaneous bleeding, delayed wound healing, and unusual scar formation; increased
incidence of spontaneous abortion
Laboratory

✓ 5.0 M urea test abnormal, PT and aPTT normal, enzymatic and immunologic studies can
be done

Acquired Disorders of Coagulation and Fibrinolysis


1. Hepatic disease

✓ The liver is the major site of hemostatic protein synthesis.


✓ Hepatic disease can result in decreased synthesis of coagulation or regulatory proteins;
it also causes impaired clearance of activated hemostatic components.

Laboratory:

✓ Prolonged PT, aPTT, bleeding time, and possibly decreased platelet counts because of
hypersplenism, alcohol toxicity, and disseminated intravascular coagulation (DIG)

2. Vitamin K deficiency

✓ Vitamin K is needed for liver synthesis of functional factors II, VII, IX, andX.
✓ Vitamin K is produced by normal intestinal flora.
✓ Deficiencies in vitamin K can result from oral antibiotics, vitamin K antagonists
(warfarin), or decreased absorption resulting from obstructive jaundice.
✓ Breast-fed babies are more prone to vitamin K deficiency because breast milk is sterile,
which allows no bacterial intestinal colonization to occur.
Laboratory:
✓ Prolonged PT (VII, X, II) and prolonged aPTT (IX, X, II)

3. Disseminated intravascular coagulation with secondary fibrinolysis


✓ Predisposing condition triggers systemic clotting; leads to systemic fibrinolysis and
bleeding
✓ Triggering events include gram-negative septicemia, acute promyelocytic leukemia (FAB
M3), obstetrical complications, massive tissue damage.
✓ Fibrinogen group factors (I, V, VIII, XIII) and platelets are consumed in clotting.
Laboratory
✓ PT, aPTT, and thrombin time are prolonged.
✓ Platelet count, antithrombin, and fibrinogen concentrations are decreased.
✓ Fibrin and fibrinogen degradation products are present (abnormal D-dimer and FDP
tests).
✓ Schistocytes form when RBCs are fragmented by intravascular clots
✓ Schistocytes form when RBCs are fragmented by intravascular clots.
Clinical:
✓ A systemic thrombotic event causes multiple organ failure; systemic lysis ultimately
leads to severe hemorrhage.

Treatment:
✓ Treat the underlying condition with FFP, platelet transfusions, antithrombin
concentrates, and heparin to stop systemic clotting.

4.Primary fibrinogenolysis
✓ Plasminogen is inappropriately activated to plasmin in the absence of clot formation.
Plasmin circulates free in plasma and destroys factors I, V, and VIII.
✓ Caused by certain malignancies (e.g., prostate cancer) or massive tissue damage that
causes release of plasminogen activators
Laboratory
a. PT, aPTT, and thrombin time are prolonged, and fibrinogen concentration is low
(plasmin degrades fibrinogen, V, and VIII).
b. Platelet count, RBC morphology, and antithrombin concentration are normal because
there is no clot formation.
c. Fibrinogen degradation products are present (abnormal FDP test), but fibrin
degradation products are absent (normal D-dimer because there is no clot formation).

Clinical:
✓ Hemorrhagic symptoms occur that may resemble DIG.
Treatment
✓ Epsilon aminocaproic acid (EACA) is used to turn off inappropriate systemic lysis.

EVALUATION TESTS FOR THE FIBRINOLYTIC SYSTEM


A. Fibrin Degradation Products (FDPs):
✓ Latex particles are coated with antibody against fibrinogen and are mixed with patient
serum. Macroscopic agglutination indicates degradation products.
✓ This is a nonspecific test that will be abnormal when either fibrin degradation products
or fibrinogen degradation products are present (DIG and primary fibrinogenolysis).

B. D-Dimer Assay:

✓ Latex particles are coated with antibody against D-dimer


✓ Highly specific measurement for fibrin degradation products; does not detect
fibrinogen degradation products.
✓ Abnormal result indicates a clot has formed, been stabilized by factor XHIa, and is being
lysed by plasmin (abnormal in DIC, but normal in primary fibrinogenolysis)

SAMPLE COLLECTION, HANDLING, AND PROCESSING FOR COAGULATION TESTING


1. Nontraumatic Venipuncture:
✓ It is essential that trauma be avoided because it may introduce tissue thromboplastin
that would activate coagulation
2. Order of Draw:
✓ It is important that proper order of draw be followed. Collect tube for coagulation
testing before any tubes containing heparin, EDTA, sodium fluoride, or clot-promoting
additives.
3. Use Plastic- or Silicone-Coated Glass Tubes:
✓ Plain glass tubes will activate the intrinsic pathway, including the activation of the
contact factors prekallikrein, XI, and XII.
4. Ratio of Blood to Anticoagulant:
✓ The ratio in blood collection tubes is critical, and it must be maintained at a 9:1 ratio of
blood to 3.2% sodium citrate anticoagulant or excess citrate will bind calcium chloride in
the reagents for PT and aPTT, causing falsely long coagulation times.
5. Specimen Processing
✓ Specimens must be processed as soon as possible following blood collection.
Recommendations include processing within 4 hours for aPTT and 24 hours for PT.
Centrifuge to obtain platelet-poor plasma, and remove plasma from cells; can freeze
plasma at -20°C.
6. Temperature:
✓ Testing must be performed at 37°C. Enzyme reactions work best at 37°C. Labile factors V
and VIII will break down at temperatures above 37°C. Factors VII and XI will be activated
at cold temperatures

ANTICOAGULANT THERAPIES

A. Unfractionated Heparin Therapy


✓ Treatment of choice to prevent extension of existing clots due to acute thrombotic
events (e.g., venous and arterial thrombosis, pulmonary embolism, thrombophlebitis,
acute myocardial infarction)
✓ Therapy involves a bolus of heparin, followed by continuous infusion.
✓ Antithrombin must be present with levels of 40-60% of normal for heparin to work.
✓ The antithrombin/heparin complex inhibits serine proteases, including Xlla, XIa, IXa, Xa,
Ha, and kallikrein. Inhibition is immediate.
✓ It inhibits the conversion of fibrinogen to fibrin, platelet aggregation, and activation of
factor XIII.
✓ Heparin activity can be immediately reversed by administration of protamine sulfate.
✓ Monitor with aPTT; therapeutic range is approximately 1.5-2 times patient's baseline
aPTT value prior to treatment. Dosage is adjusted accordingly.
✓ Daily platelet counts should be performed on heparinized patients to monitor for
heparin-induced thrombocytopenia (HIT). If detected, heparin therapy is immediately
halted and different anticoagulant therapies are considered.

B. Warfarin (Coumadin®/Coumarin) Therapy


✓ This oral anticoagulant is prescribed on an outpatient basis to prevent extension of
existing clots and recurrence of thrombotic events, and prophylactically it is often
prescribed post surgery to prevent thrombosis.

✓ Vitamin K antagonist 3. Warfarin inhibits liver synthesis of functional prothrombin


group factors II, VII, IX, and X. Factor VII is affected first (short half-life) and to the
greatest extent

c. Antiplatelet medications
✓ Like aspirin, Plavix®, ticlopidine, and nonsteroidal anti-inflammatory drugs/NSAIDS
✓ may be used in conjunction with other anticoagulant therapies to prevent recurrence of
thrombotic events

You might also like