You are on page 1of 158

MODULE 1: Introduction to Immunohematology (part1)

MLS 18, February 23, 2021


BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
1628 William Harvey- Discovered the circulation of blood.
Legend:
The first recorded successful blood transfusion occurred in
Transcription Bullet 1665
England.
Notes/Module Packet Bullet
Jean Denis - Published in the Philosophical Transactions his
Reference Book 1667
experience in Paris with transfusing lamb blood.
Highlighted box- High yield information
Dr. James Blundell- The first to successfully transfuse
1818-
MODULE OUTLINE human blood into a patient with post-partum hemorrhage.
I. Historical Background and the current trends in Transfusion Samuel Armstrong Lane- Performs the first successful
1840
Medicine whole blood transfusion to treat hemophilia.
Joseph Lister- Uses antiseptics to control infection during
II. Blood Preservation 1867
transfusions.
III. Review of Basic Genetics US physicians transfuse milk (from cows, goats, and
IV. Review of Fundamentals of Immunology 1873-1880
humans)
V. Review of Molecular Biology Saline infusion replaces milk as a “blood substitute” due to
1884
the increased frequency of adverse reactions to milk.
LEARNING OUTCOMES  Blood was taken from 3 young men; unfortunately, all four
1. List the major developments in the history of transfusion medicine. (including the Pope) died.
o Clottin was the principal obstacle to overcome.
o Attempts to find non-toxic anticoagulant began in 1868, when
Braxton Hicks recommended sodium phosphate.
HISTORICAL BACKGROUND AND THE CURRENT TRENDS IN
o This was perhaps the first example of blood preservation research
TRANSFUSION MEDICINE
o Human blood transfusion is the process of transferring blood or BLOOD GROUPS
blood-based products from an individual into the circulatory system
of another.
1901- Edward Lindeman
o Over the past 100 years, blood transfusion has grown from the
transfusion of small amounts of fresh whole blood, to one of the o The first to succeed in designing a device for transfusion.
most common therapeutic medical practices. o Performed a vein-to-vein blood transfusion by using multiple
syringes & a special canula for transfusion from donor to
EARLY TRANSFUSION patient.

1492- Pope Innocent VII


o First blood transfusion recorded

#MLSWATER2022 Page 1 of 4
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
Audrey Smith- Reported the use of glycerol cryoprotectant
Karl Landsteiner- Discovered the first three human blood 1950
for freezing red blood cells.
1900 groups, A, B, and C. (Blood type C was later changed to O.) Gibson et al developed citrate-phosphate-dextrose
(CPD). CPD eventually replaced ACD and became
Alfred Decastello and Adriano Sturli- Added AB as the
1957 commonly used preservative for storage of blood/red
1902 fourth human blood group.
cells in liquid form. Shelf-life of blood stored in CPD at
Hektoen- Suggested that the safety of transfusion might be 2-4 °C was 21 day.
improved by crossmatching blood between donors and A new anticoagulant preservative, CPDA-1, extends the
patients to exclude incompatible mixtures. Reuben shelf life of whole blood and red blood cells to 35 days,
Ottenberg performed the first blood transfusion using increasing the blood supply and facilitating resource sharing
1907 1979
blood typing and crossmatching in New York. Ottenberg among blood banks. The addition of adenine improved the
also observed the mendelian inheritance of blood groups synthesis of ATP in the stored blood, which prolonged the
and recognized the “universal” utility of group O donors. storage of blood/red cells at 2-4 °C to 35 days.

1908 Moreschi- Described the antiglobulin reaction. 1941- Dr. Charles Drew
Dr. Karl Landsteiner- published the first of a series of o Developed techniques on blood transfusion and blood preservation
1990 papers demonstrating presence of the ABO blood group
during WWII
system.
o Led to the establishment of a widespread system of blood banks.
Landsteiner, Weiner, Levine and Stetson- Discovered the
1939-1940 o Director of the first American Red Cross blood bank at
Rh blood group system.
1927-1947 The MNSs and P systems were discovered Presbyterian Hospital
Coombs, Mourant, and Race- Described the use of
1945 antihuman globulin sera to detect IgG antibodies in
compatibility testing.
BLOOD STORAGE
BLOOD DERIVATIVES
1908 Alexis Carrel- Performed direct transfusion.
Edwin Cohn- Developed the cold ethanol fractionation
Long-term anticoagulants, among them sodium citrate, 1940
1914 process, called Cohn fractionation.
were developed.
Pool and Shannon- Revolutionized the treatment of
Rous and Turner- Used sodium citrate as an anticoagulant 1961
1916 hemophilia A.
with glucose.
Concentrated Rh immune globulin was introduced
1932 The first blood bank was established in a Leningrad hospital 1967
commercially.
Bernard Fantus- Established the first hospital blood bank in
1937 Dry-heated, lyophilized factor VIII and IX concentrates
the United States. 1985
became available.
Loutit and Mollison- Developed acid citrate dextrose (ACD)
1943 Genetically engineered (recombinant) factor VIII became
solution. 1993
available.
1998 Factor IX became available
#MLSWATER2022 Page 2 of 4
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
A test for the HIV antibody was introduced; FDA approves
BLOOD COMPONENT THERAPY 1985 enzyme-linked immunosorbent assay (ELISA), first blood-
screening test to detect HIV antibodies.
Roger Lee and Paul Dudley White- Developed the Lee- Two tests that screen for indirect evidence of hepatitis
1912
White clotting time. were developed and implemented, hepatitis B core
1987
Walter and Murphy- Introduced the use of plastic bags as a antibody (anti-HBc) and the alanine aminotransferase test
1950 (ALT).
replacement for glass bottles.
Concentrated blood platelets were recognized as useful for Testing of donated blood for human-Tlymphotropic-virus-I-
1961 1989
the treatment of thrombocytopenia. antibody (anti-HTLV-I) begins.
The first antihemophilic factor (AHF) concentrate to treat 1990 Testing for hepatitis C became routine.
1962 coagulation disorders in hemophilia patients was 2002 West Nile virus identified as transfusion transmissible.
developed through fractionation.
Plasmapheresis was introduced as a means of collecting What are the different obstacles that they need to overcome in order
1964 to have a successful blood transfusion?
plasma for fractionation.
Rh immune globulin was commercially introduced to o Problem with the clotting
1967 prevent Rh disease in the newborns of Rh-negative o Use of Anticoagulant
o Compatibility Testing
women.
o Antiseptic Techniques
Hepatitis B surface antigen (HBsAg) testing of donated
1971 o Blood Preservation
blood begins. o Component Therapy- in cases of Transfusion Overload
1972 Platelets for transfusion were collected by apheresis.

ADVERSE EFFECT OF TRANSFUSION ADVANCES IN BLOOD TRANSFUSION


Today, the blood transfusion community continues to advance its
Greenwalt et al.- Demonstrated leukocyte reduction filters transfusion systems, guided by the:
1962-
prevented febrile reactions. o ISBT (International Society of Blood Transfusion),
1970- Graw et al.- Used irradiation to prevent TA-GVHD. o AABB (American Association of Blood Banks),
Commercial testing for hepatitis B surface antigen began. o FDA (Food and Drug Administration), and
1971-
o Other federal and professional organizations.
First Acquired Immune Deficiency Syndrome (AIDS) case Researchers are establishing new surveillance systems that record
1981
was reported. data and transfusion outcomes (haemovigilance system).
1983- Transfusion-transmitted HIV was described. o They are offering more personalized treatment, limiting
Human Immunodeficiency Virus (HIV) was identified asthe transfusions based on careful assessment of need, and
1984 ultimately improving patient care.
cause of AIDS.
In addition to infectious disease risks, treating physicians must also
manage other risks, such as post-transfusion reactions.

#MLSWATER2022 Page 3 of 4
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o These include:  Variant Creutzfeldt-Jakob
 Transfusion-related lung injury (TRALI);  West Nile Virus
 Transfusion associated cardiac overload (TACO); and  Malaria
 Post-transfusion iron overload.  Babesiosis
To minimize these risks, researchers studying the body’s immune  Chagas Disease
response to transfusions have found that modifying the blood prior to
transfusion can reduce reactions. STEP 3: The Abbreviated Physical Examination
o In particular, removing white blood cells or radiating blood. o The abbreviated physical examination for donors includes;
o Recently, studies found that using male plasma and platelets  Blood pressure, Pulse, and Temperature readings;
may eliminate the transmission of certain antibodies that can  Hemoglobin or Hematocrit level;
cause reactions and are found only in previously pregnant  Inspection of the arms for skin lesions
women and transfused males.
Over the last 15 years, we’ve seen a shift from whole blood–derived Currently 10 screening tests for infectious disease are performed on
platelets obtained through whole-blood collection to single-donor each unit of donated blood (See Table 1-1; Harmening, 6th Edition)
platelets obtained through apheresis.
o Blood components collected by apheresis offer several
-END OF TRANSCRIPTION-
advantages over manually collected whole blood.
o Because apheresis separates the components, the blood
doesn’t need further processing.
Many apheresis devices allow collection of plasma and RBCs
simultaneously with platelets. Although components still need to be
tested, labeled, and stored appropriately, apheresis eliminates one
processing step. Some apheresis devices also leukoreduce RBCs and
platelets, eliminating the need to filter them in the laboratory.

DONATION PROCESS
STEP 1: Educational Materials
o Educational (ex. AABB pamphlet “An Important Message to Blood
Donors”) that contains information on the risks of infectious diseases
transmitted by blood transfusion, including the symptoms and signs of
AIDs, is given to each prospective donor to read.

STEP 2: The Donor Health History Questionnaire


o A uniform donor history questionnaire, designed to as questions that
protect the health of both the donor and the recipients, is given to
every donor.
o The health history questionnaire is used to identify donors who have
been exposed to diseases that can be transmitted in blood
 Example of blood-transmitted diseases;
#MLSWATER2022 Page 4 of 4
MODULE 1: Introduction to Immunohematology (part 2)
MLS 18, February 23, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
RBC BIOLOGY
Legend: Three areas of RBC biology are crucial for normal erythrocyte survival
Transcription Bullet and function:
Notes/Module Packet Bullet 1. Normal chemical composition and structure of the RBC membrane
Reference Book 2. Hemoglobin structure and function
MODULE OUTLINE 3. RBC metabolism Defects in any or all of these areas will result in
I. Historical Background and the current trends in Transfusion Medicine RBCs surviving less than the normal 120 days in circulation.
II. Blood Preservation
Normal chemical composition and structure of the RBC membrane
A. RBC Biology
B. Blood Preservation RBC MEMBRANES
III. Review of Basic Genetics
Phospholipids
IV. Review of Fundamentals of Immunology
o The main lipid components of the membrane, are arranged in a
V. Review of Molecular Biology bilayer structure comprising the framework in which globular
proteins traverse and move.
LEARNING OUTCOMES
1. Describe the biologic properties of red blood cells (RBC) that can Integral Membrane Proteins
affect post-transfusion survival. o Proteins that extend from the outer surface and span the entire
2. Identify the metabolic pathways that are essential for normal RBC membrane to the inner cytoplasmic side of the RBC
function & survival.
3. Explain the importance of 2-3 DPG levels in transfused blood.
Peripheral Proteins
4. List the approved anticoagulant preservative solutions, explain the
o Second class of membrane proteins, found beneath the lipid
function of each component, and state the maximum storage time
bilayer
for RBCs and platelets collected in each.
o located and limited to the cytoplasmic surface of the membrane
5. Explain how additive solutions are used and list their advantages.
forming the RBC cytoskeleton
6. Explain storage requirements for platelets, including rationale.
7. Describe the indications for platelet transfusion.
8. Explain the importance of platelet corrected count increment (CCI). Integral Proteins Peripheral Proteins
Glycophorin A Spectrin
Glycophorin B Actin (band 5)
Glycophorin C Ankyrin (band 2.1)
Anion-exchange-channel protein Band 4.1 and 4.2
(band 3) Band 6
Adducin
Table 1: RBC Membrane Integral and Peripheral Proteins
#MLSWATER2022 Page 1 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
NOTE: 1. Deformability
Red blood cells are a product of a differentiation process that starts To remain viable, normal RBCs must also remain flexible, deformable,
in the bone marrow where hematopoietic stem cells differentiate to and permeable.
nucleate RBCs. After extrusion of nuclei and degradation of The loss of adenosine triphosphate (ATP) (energy) levels leads to a
endoplasmic reticulum, reticulocytes emerge in the circulation; here decrease in the phosphorylation of spectrin and, in turn, a loss of
they rapidly develop into mature RBCs. membrane deformability.
Despite these features, the protein and lipid composition of the RBCs An accumulation or increase in deposition of membrane calcium
is subject to change during its life-time. This can be particularly also results, causing an increase in membrane rigidity and loss of
observed at the level of its plasma membrane. pliability.
Changes in red blood cell morphology occurred as quickly as 22 days. o These cells are at a marked disadvantage when they pass through
o This alteration can be harmful because red blood cells are similar in the small (3 to 5 μm in diameter) sinusoidal orifices of the
size to the diameter of small capillaries; therefore, red blood cells spleen, an organ that functions in extravascular sequestration and
have to change shape to get through the capillaries. removal of aged, damaged, or less deformable RBCs or fragments
of their membrane.
The loss of RBC membrane is exemplified by the formation of
“spherocytes” (cells with a reduced surface-to-volume ratio) and
“bite cells,” in which the removal of a portion of membrane has left a
permanent indentation in the remaining cell membrane.
o The survival of these forms is also shortened.

2. PERMEABILITY
The permeability properties of the RBC membrane and the active
RBC cation transport prevent colloid hemolysis and control the
volume of the RBC.
o Any abnormality that increases permeability or alters
cationic transport may decrease RBC survival.
The RBC membrane is freely permeable to water and anions.
Figure 1. Schematic illustration of red blood cell membrane depicting the composition
Chloride (Cl–) and bicarbonate (HCO3–) can traverse the membrane
and arrangement of RBC membrane proteins.
in less than a second.
The RBC membrane is relatively impermeable to cations such as
RBC CHARACTERISTICS sodium (Na+) and potassium (K+).
The normal chemical composition and the structural arrangement RBC volume and water homeostasis are maintained by controlling
and molecular interactions of the erythrocyte membrane are crucial the intracellular concentrations of sodium and potassium.
to the normal length of RBC survival of 120 days in circulation. In o The erythrocyte intracellular-to-extracellular ratios for Na+
addition, they maintain a critical role in two important RBC and K+ are 1:12 and 25:1, respectively.
characteristics: deformability and permeability.
The 300 cationic pumps, which actively transport Na+ out of the cell
and K+ into the cell, require energy in the form of ATP.
Calcium (Ca2+) is also actively pumped from the interior of the RBC
through energy-dependent calcium-ATPase pumps. Calmodulin, a
#MLSWATER2022 Page 2 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
cytoplasmic calcium-binding protein, is speculated to control these “Tense” Form
pumps and to prevent excessive intracellular Ca2+ buildup, which o The resulting conformation of the deoxyhemoglobin molecule is
changes the shape and makes it more rigid. known as the tense (T) form, which has a lower affinity for
When RBCs are ATP depleted, Ca2+ and Na+ are allowed to oxygen.
accumulate intracellularly, and K+ and water are lost, resulting in a
dehydrated rigid cell subsequently sequestered by the spleen, “Relaxed” Form
resulting in a decrease in RBC survival. o When hemoglobin loads oxygen and becomes oxyhemoglobin,
the established salt bridges are broken, and chains are pulled
HEMOGLOBIN STRUCTURE AND FUNCTION together, expelling 2-3-DPG. This is the relaxed (R) form of the
hemoglobin molecule, which has a higher affinity for oxygen.
HEMOGLOBIN
Hemoglobin is the major protein in RBCs, and it gives to red cells the
ability to transport oxygen and carbon dioxide to and from tissues. HEMOGLOBIN-OXYGEN DISSOCIATION CURVE
These allosteric changes that occur as the hemoglobin loads and
unloads oxygen are referred to as the respiratory movement.
HEMOGLOBIN FUNCTION
Are not directly proportional to the partial pressure of oxygen (pO2)
Hemoglobin’s primary function is gas transport: in its environment but instead exhibit a sigmoid-curve relationship,
o Oxygen (O2) delivery from the lungs to the tissues known as the hemoglobin-oxygen dissociation curve.
o Transport of Carbon dioxide (CO2) from tissues to the lungs The shape of this curve is very important physiologically because it
permits a considerable amount of oxygen to be delivered to the
2,3-DPG tissues with a small drop in oxygen tension.
One of the most important controls of hemoglobin affinity for o For example, in the environment of the lungs, where the
oxygen is the RBC organic phosphate 2,3-DPG. oxygen (pO2) tension, measured in millimeters of mercury
A carbon molecule important in erythrocyte metabolism. (mmHg), is nearly 100 mm Hg, the hemoglobin molecule is
o It binds to deoxygenated hemoglobin and increases oxygen off- almost 100% saturated with oxygen.
loading from hemoglobin into the tissues. As the RBCs travel to the tissues, where the (pO2) drops to an
o As erythrocyte storage time increases, the levels of 2,3-DPG average of 40 mm Hg (mean venous oxygen tension), the
decrease. hemoglobin saturation drops to approximately 75% saturation,
 Transfusion of 2,3-DPG-depleted blood may shift the releasing about 25% of the oxygen to the tissues.
oxygen–hemoglobin dissociation curve to the left. o This is the normal situation of oxygen delivery at a basal
 As a result, red blood cells will have difficulty in unloading metabolic rate.
oxygen from hemoglobin into the tissues. Ligands
 The unloading of oxygen by hemoglobin is accompanied by o The normal position of the oxygen dissociation curve depends
widening of a space between chains and the binding of 2,3- on three different ligands normally found within the RBC: H+
DPG on a mole-for-mole basis, with the formation of anionic ions, CO2, and organic phosphates.
salt bridges between the chains. o Of these three ligands, 2,3-DPG plays the most important
physiological role. Normal hemoglobin function depends on
adequate 2, 3-DPG levels in the RBC.

#MLSWATER2022 Page 3 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
 In situations such as hypoxia, a compensatory “shift to the RBC Metabolism
right” of the hemoglobin-oxygen dissociation curve RBCs produce ATP anaerobically by the breakdown of glucose, thus
alleviates the tissue oxygen deficit. This rightward shift of not using any of the oxygen for its own metabolism.
the curve, mediated by: o Erythrocytes do not have mitochondria, which is the site of
 Increased levels of 2,3-DPG; aerobic respiration.
 Decreases hemoglobin’s affinity for the oxygen
Anaerobic metabolism allows red blood cells to deliver 100% of the
molecule and;
oxygen to the organ sites. RBC metabolism may be divided into the
 Increases oxygen delivery to the tissues.
anaerobic glycolytic pathway and three ancillary pathways that
serve to maintain the structure and function of hemoglobin:
 A “shift to the left” of the hemoglobin-oxygen dissociation
1. The Pentose Phosphate pathway
curve results, conversely;
2. The Methemoglobin Reductase pathway
 Increase in hemoglobin oxygen affinity and a decrease
3. The Luebering-Rapoport shunt.
in oxygen delivery to the tissues.
All of these processes are essential if the erythrocyte is to transport
 With such a dissociation curve, RBCs are much less
oxygen and to maintain critical physical characteristics for its
efficient because only 12% of the oxygen can be
survival.
released to the tissues.
o Glycolysis generates about 90% of the ATP needed by the RBC.
 Multiple transfusions of 2,3-DPG–depleted stored blood
o Approximately 10% is provided by the pentose phosphate
can shift the oxygen dissociation curve to the left.
pathway.
o The methemoglobin reductase pathway is another important
pathway of RBC metabolism, and a defect can affect RBC post-
transfusion survival and function.
RBC metabolism includes the glycolytic pathways producing both
energy (as adenosine 5′- triphosphate, or ATP) and oxidation-
reduction intermediates that support oxygen transport and
membrane flexibility. RBCs interact with their environment by
changing shape in response to pH and by secreting ATP in response
to sheer forces and nitric oxide in response to hypoxia.
At the end of their life span, RBCs undergo programmed cell death
in two ways:
o By racemization of negatively charged membrane phospholipids
in response to calcium or low concentrations of ATP, and
o By the active loss of membrane through microvesiculation.
Figure 1. Diagrammatic representation of oxygen equilibrium curves of the
lug worm, man, and hemoglobin Scuba. Storage Lesion
o The effect of hydrogen ions, 2, 3-bisphosphoglycerate, and carbon o A blanket term used to encompass all of the “bad” things that
dioxide (H++BPG +CO2) is to promote a right shift. happen to RBCs during storage. These “bad” things include:
o If man had the hemoglobin of the lug worm (left shift), he would die  Decreased concentrations of ATP and 2,3-
of anoxia. diphosphoglycerate (2,3-DPG)

#MLSWATER2022 Page 4 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
 Increased concentrations of extracellular potassium, o Two molecules of lactic acid contain exactly the same number of
changes in cell shape carbons, hydrogens, and oxygens as one molecule of glucose;
 Loss of RBC viability  there is sufficient free energy available from the cleavage
 Hemolysis and rearrangement of the glucose molecule to produce 2
moles of ATP per mole of glucose converted into lactate.
CLINICAL SIGNIFICANCE The RBC uses most of this ATP to maintain electrochemical and ion
1. Elevated extracellular potassium concentrations in stored RBC units gradients across its plasma membrane.
Not usually of clinical significance One mole of glucose is converted to 2 moles of lactate during
However, can be fatal if RBC concentrates are infused in large anaerobic glycolysis.
volumes through central vascular catheters No oxygen is consumed, nor is CO2 produced in this pathway. There
o As in neonatal exchange transfusions or when priming is a net yield of 2 mole of ATP per mole of glucose converted to
cardiopulmonary bypass machines. lactate.

2. Hemolysis LUEBERING-RAPAPORT PATHWAY


Reduction of the relative number of stored RBCs that survive
An offshoot of the Embden-Meyerhof pathway is the Luebering-
when returned to circulation
Rapaport bypass.
Release of harmful breakdown products.
This pathway permits the accumulation of an important RBC organic
phosphate, 2,3-diphosphoglycerate (2,3- DPG).
3. Free hemoglobin
o Has a significant effect on the affinity of hemoglobin for oxygen
Vasoconstriction
and therefore affects how well RBCs function post-transfusion.
Endothelial cell activation
Renal tubular damage. The 2,3- DPG alternate route does not yield ATP, but does modulate
levels of 2,3-DPG.
4. Free membrane phospholipids Erythrocyte 2,3-DPG concentration is especially sensitive to pH
Are procoagulant because the enzymes catalyzing its production are inhibited or
Clinical consequences of the early forms of RBC shape change or stimulated by hydrogen ions.
reduced 2,3-DPG concentrations are much less clear. Defects in the Luebering-Rapaport bypass can affect the levels of
2,3-DPG available to erythrocytes. Red blood cell 2,3-DPG regulates
5. Loss of viability during storage oxygen release depending on the needs of tissues.
Reduces the effective transfused RBC dose, but the Whenever the peripheral tissues have an increased amount of
consequences of administering intact but nonviable cells are not deoxygenated blood (deoxyhemoglobin), glycolysis is stimulated
known. and 2,3-DPG levels rise.
o Again, pH changes within the cell probably contribute to this
ANAEROBIC GLYCOLYTIC PATHWAY process.
o The result is that 2,3- DPG attaches to deoxyhemoglobin and
In RBCs, which lack mitochondria and oxidative metabolism,
causes hemoglobin to resist binding to oxygen. This decrease in
pyruvate is reduced to lactic acid, a three-carbon hydroxyacid, the
oxygen affinity by hemoglobin increases oxygen release to
product of anaerobic glycolysis.
tissues.
Each mole of glucose yields 2 moles of lactate, which are then
excreted into blood.
#MLSWATER2022 Page 5 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
HEXOSE MONOPHOSPHATE SHUNT/ PATHWAY component of hemoglobin, the methemoglobin reductase pathway
Red blood cells are capable of limited aerobic glycolysis ensures that the iron (heme) in the hemoglobin molecule does not
Also called the phosphogluconate pathway or the pentose become oxidized. Methemoglobin with iron in the ferric state is
phosphate shunt. useless as an oxygen carrier. This pathway uses the enzyme
methemoglobin reductase and NAD to maintain hemoglobin in a
The major role of the hexose monophosphate shunt, is the
reduced state.
generation of reduced nicotinamide adenine dinucleotide
phosphate (NADPH).
o Erythrocyte NADPH converts oxidized glutathione to reduced THE IMPORTANCE OF 2-3 DPG LEVELS IN TRANSFUSED BLOOD
glutathione, the major red blood cell antioxidant. The two main regulators of oxygen uptake and delivery are the pH
o Red blood cell enzymes, and especially hemoglobin, are of tissues and the content of 2,3-diphosphoglycerate (2,3-DPG) in
protected from oxidant damage through the action of red cells.
glutathione, which maintains hemoglobin in a reduced, active The pH of blood is kept relatively constant at the slightly alkaline
form. level of about 7.4 (pH less than 7 indicates acidity, more than 7
Although oxidants are damaging to cells, cells commonly produce alkalinity). The effect of pH on the ability of hemoglobin to bind
them. oxygen is called the Bohr effect:
o Macrophages, for example, produce them in response to o When pH is low, hemoglobin binds oxygen less strongly, and
infection. when pH is high (as in the lungs), hemoglobin binds more tightly
o Erythrocytes even produce them when certain drugs are present. to oxygen.
If the level of reduced glutathione is not sufficient to neutralize o The Bohr effect is due to changes in the shape of the hemoglobin
intracellular red blood cell oxidants, globin will denature and molecule as the pH of its environment changes. The oxygen
precipitate as Heinz bodies, ultimately producing membrane affinity of hemoglobin is also regulated by 2,3-DPG, a simple
damage. The levels of nicotinamide adenine dinucleotide phosphate molecule produced by the red cell when it metabolizes glucose.
(NADP)/NADPH regulate the amount of glucose metabolized by the Effect of 2,3-DPG is to reduce the oxygen affinity of hemoglobin.
hexose monophosphate shunt. When the availability of oxygen to tissues is reduced, the red cell
As NADPH generates reduced glutathione, NADP is produced, which responds by synthesizing more 2,3-DPG, a process that occurs over
stimulates glucose metabolism in the hexose monophosphate a period of hours to days. 2,3-DPG is an intermediary metabolite in
shunt. This mechanism arms the red blood cell with more reducing the Embden–Meyerhof glycolytic pathway in the red cells, which
capability during an oxidative challenge. affects haemoglobin affinity for oxygen.

↑ Concentration of 2,3-DPG ↓ Concentration of 2,3-DPG


METHEMOGLOBIN REDUCTASE PATHWAY
o decreases the affinity and thus o Has the opposite effect.
An important auxiliary process of erythrocyte metabolism is the increases the fraction of o Caused by acidosis.
methemoglobin reductase pathway. haemoglobin-bound oxygen
This pathway maintains heme iron in the reduced, or active state available to the tissues
(ferrous). o found in response to hypoxia or
It requires reduced pyridine nucleotides generated from the anemia
Embden-Meyerhof pathway.
Unlike the hexose monophosphate shunt, which provides a Table 2: Effects of 2,3-DPG
mechanism for preventing the denaturation of the globin

#MLSWATER2022 Page 6 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
BLOOD PRESERVAT ION ANTICOAGULANT PRESERVATIVE SOLUTIONS IN BLOOD BAGS
The goal of blood preservation is to provide viable and functional Prevent blood from clotting
blood components for patients requiring blood transfusion. Also used to preserve the life and survival of RBCs so as to have the
maximum post transfusion survival.
RBC Viability Citrate-based anticoagulants are used for the collection of blood
A measure of in vivo RBC survival following transfusion. for transfusion.
Because blood must be stored from the time of donation until the o Citrate binds calcium, preventing the activation of plasma
time of transfusion, the viability of RBCs must be maintained during coagulation factors.
the storage time as well. EDTA, which binds calcium more strongly compared to citrate, is not
To maintain optimum viability, blood is stored in the liquid state used for the collection of blood transfusion products.
between 1°C and 6°C for a specific number of days, as determined Heparin is rarely used for the collection of blood transfusion
by the preservative solution(s) used. components as its effective anticoagulant half-life (approximately
2,3-DPG is re-formed in stored RBCs after in vivo circulation an hour) is limited.
Whole blood is collected into a primary blood bag containing a
Factors affecting the rate of 2,3-DPG restoration citrate based anticoagulant (CPD, CPDA-1 or CP2D) and adequate
o Recipient’s Acid-Base status mixing is ensured. The anticoagulant solution may contain nutrients
o Phosphorus Metabolism such as glucose and adenine.
o Degree of Anemia Additional cell specific nutrients are supplied to the stored red cells
o Overall severity of the disorder and platelets by the additive solutions in the respective red cell and
RBC clearance occurs within the first hour after transfusion platelet satellite bags.
Approximately 220 to 250 mg of iron in one RBC unit Citrate is the most common anticoagulant used in apheresis blood
Rapid RBC clearance of a single unit of blood delivers a massive load collection. Sodium citrate (4%) is used for plasma apheresis and
of iron to the monocyte and macrophage system ACD-A is used for platelet apheresis.
Incorporation of Adenine & its effects on glycolysis and ATP levels
Food and Drug Administration requirements o Adenine incorporated into the CPD solution (CPDA-1) increases
1. Average 24-hour post transfusion RBC survival of more than 75% ADP Levels, thereby driving glycolysis toward the synthesis of
ATP.
2. Free Hemoglobin less then 1% of total hemoglobin
NAME ABBREVIATION STORAGE
Determination of Post-Transfusion RBC survival TIME (DAYS)
 Taking RBCs from healthy subjects Acid -citrate-dextrose ACD 21
 Storage of RBCs labeled with radioisotopes Citrate-Phosphate-Dextrose CPD 21
 Reinfusion back to the original donor Citrate-Phosphate-Dextrose-Adenine CPDA-1 35
 Measurement 24 hours after transfusion Citrate-Phosphate-Double-Dextrose CP2D 21
Table 3: Additive Solutions in use in North America

#MLSWATER2022 Page 7 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
Pathophysiologic effects of the transfusion of RBCs with low 2,3- CITRATE PHOSPHATE DEXTROSE ADENINE (CPDA-1)
DPG levels & increased affinity for Oxygen: CPD to which adenine (A) is added, becomes CPDA-1 (the‘1’
o ↑ Cardiac Output signifies the formula used) and improves the synthesis of ATP.
o ↓Mixed venous (pO2) tension CPDA-1 is usually used when the collected donation is to be stored
o ↑Cardiac output & ↓pO2 tension as whole blood.

Plastic containers used for storage of blood affects its viability NOTE:
di (ethylhexyl)-phthalate (DEHP) in PVC bags The volume of anticoagulant required to prevent clotting and
o Plasticizer preserve red cells is dependent on the volume of blood taken
o Leach from the plastic into the lipids of the plasma medium and from the donor.
RBC membranes of the blood during storage Some collection bags are designed for the collection of 500 mL
o Break at low temperature blood and contain 70 mL anticoagulant; others are designed for
Added to the RBCs after removal of the plasma 450 mL collections and contain 63 mL anticoagulant.
Without ADSOL: If smaller quantities of blood are to be drawn, then the volume
o Decreased nutrients necessary to maintain RBCs during of anticoagulant is reduced proportionately. 15 ml of ACD, 14ml
storage of CPD or CPDA is used for preserving 100ml of blood.
o Decreased in viability, particularly in the last 2 weeks of
storage.
ACTION OF INGREDIENTS OF ANTICOAGULANT SOLUTION
ACID CITRATE DEXTROSE (ACD):
The original ACD solution had a pH of 5; CITRATE Acts by chelating Calcium.
Was made with citric acid, dextrose, and sodium citrate; and stably DEXTROSE Necessary for the metabolism of stored RBCs. It passes
survived being autoclaved. from plasma into the red cells and is utilized for energy
It was used primarily for glass bottle storage of whole blood and production. The principal pathway being Anaerobic
allowed for 72 % RBC survival after 21 days with little hemolysis. glycolysis.
CITRIC ACID Prevents carmalization of glucose in citrate dextrose
CITRATE PHOSPHATE DEXTROSE (CPD) solution during autoclaving.
(CPD) Citrate–phosphate–dextrose (CPD), is the mainstay of blood ADENINE Improves the viability of red cells.
preservation. CPDA – 2 Here, the amount of Adenine is increased to 0.55g and
Citrate works as an anticoagulant by binding to and inhibiting the that of dextrose to 44.6g.
function of calcium (factor IV). o This is a better anticoagulant preservative
Phosphate stabilizes pH which maintains proper levels of 2,3-DPG. solution than CPDA–1.
The dextrose component is necessary for red blood cell ATP
production.
If adenine, a purine nucleotide, is added to CPD (CPD-A), storage
time jumps from 21 days to 35 days.
o Adenine assists in the production of ATP.

#MLSWATER2022 Page 8 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
STORAGE TEMPERATURES anticoagulated so the presence of CPD is no longer required by
Collection of blood into anticoagulant-preservatives maintains the red cell concentrate remaining in the primary collection bag.
component function and viability only if storage is within the However, the red cells need nutrients to survive, and should also
be suspended in sufficient fluid to allow for normal flow
correct temperature range.
characteristics. This is achieved by the use of additive solutions.
Low temperature storage slows glycolytic activity and allows
the shelf life of the component to be extended.
Low temperatures also retard bacterial proliferation, should Additive solutions (AS) vary in composition depending on the
bacteria have gained access during the time of donation; supplier. They are sometimes referred to by their brand names or
either from the venipuncture site, the donor’s circulation or simply as SAGM (saline, adenine, glucose and mannitol) or AS-1, AS-
other source. 3, AS-5 and so on. The typical composition of an additive solution is
as follows:
Additive Solution
Additive solutions are preserving solutions that are added to the SALINE the fluid in which the red cells are
RBCs after removal of the plasma with/without platelets. Additive suspended to provide the desired flow
solutions replace nutrients lost when the plasma is removed from rate conditions.
red blood cells. When additive solutions are added, red blood cell’s GLUCOSE (OR DEXTROSE) Provides the basic nutrients for glycolysis.
storage time can be increased from 35 days to 42 days. ADENINE AND MANNITOL assist in the process of ATP generation.
Two of the solutions (Adsol, Optisol) contain adenine, glucose,
saline, and mannitol. When an additive system is used then the blood donation is
o Reason for their development: removal of the plasma collected in CPD, which has no adenine.
component during the preparation of RBC concentrates removed o The unit is processed within 24 hours of collection and the
much of the nutrients needed to maintain RBCs during storage. adenine is added with the red cell additive solution.
Also overcome the problem of high viscosity of RBC o The volume of additive solution required to preserve red cells
concentrates. during storage varies according to the volume of the whole blood
With CPD anticoagulant in the primary bag, the additive solution used is donation.
SAGM (saline, adenine, glucose, mannitol). o Red cells from a donation of 500 mL require about 111 mL of
additive solution, whereas 450 mL donations need 100 mL.
ADVANTAGES Red cell concentrates and platelet concentrates are stored as fresh
Extends the storage of RBCs and lowers the viscosity of packed products ready for transfusion, although in special circumstances
red cells for ease of transfusion. Maximum amount of fresh can be frozen using specific cryopreservation procedures.
plasma is harvested – platelets and cryoprecipitate. Red cell o Red cell concentrates are stored refrigerated (2– 6 °C) for up to
concentrates that are prepared from whole blood donations 42 days (maximum duration may vary depending on the type of
collected into CPD are suspended in additive solution for additive solution used and/or local regulatory criteria).
improved storage and shelf life. o Platelet concentrates are stored at 20–24 °C for up to 5 days,
with continuous gentle agitation to maintain maximum biological
When plasma is removed after the centrifugation of whole blood function.
donations, most of the anticoagulant and nutrients in CPD are
removed along with it. At this stage, blood has been effectively
#MLSWATER2022 Page 9 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
During storage, red cells and platelets continue to metabolize and This interprets as a shelf life of 21 days for ACD, 28 days for CPD, 35
undergo a range of physiochemical changes, collectively referred to as days for CPDA-1 and 42 days for CPD replaced with a suitable additive
the 'storage lesion’. solution.
o The storage lesion ultimately affects the in vivo function and
survival of transfused red cells and platelets and thus limits their For Platelet Concentrates
shelf life. Shelf life at +22°C ± 2°C is determined by its efficacy when transfused.
This may be related to platelet viability and function during storage in
Currently approved AdSol: the correct conditions of temperature and motion, and is considered to
1. Adsol (AS-1) – Baxter Healthcare be up to 7 days.
2. Nutricel (AS-3) – Pall Corporation Most blood services allocate a 5-day shelf life to limit the increased risk
3. Optisol (AS- 5) – Teruo Corporation of bacterial growth resulting from the room temperature storage
requirement.
AS-1 AS-3 AS-5
Storage period (days) 42 42 42 For Plasma
pH (measured at 37°C) 6.6 6.5 6.5 The levels of stable clotting factors (FII, FVII, FIX, FX and fibrinogen) are
24-hour survival (%) 83 85.1 80 quite well maintained at +4°C ± 2°C.
ATP (% initial) 68 67 68.5 Labile clotting factors (FV, FVIII) deteriorate to levels that are not useful
2,3-DPG (% initial) 6 6 5 if not frozen within 24 hours of collection.
Hemolysis 0.5 0.7 0.6 Shelf life of labile coagulation factors is up to 3 years if frozen (colder
Table 4: Red Cell Additives: Biochemical Characteristics than –25°C).
However, a storage temperature that does not reach –25°C but is colder
SHELF LIFE OF WHOLE BLOOD COMPONENTS than –18°C reduces shelf life to 3 months.
SHELF LIFE is the maximum allowable storage time that a blood
product may be stored, provided that the requirements of RBC FREEZING
temperature, preservative solutions and physical environment are For Autologous Units & storage of rare blood types
met. Involves the addition of a cryoprotective agent to RBCs that are less
Because of the great diversity in collection containers and than 6 days old
anticoagulant preservatives, as well as storage capabilities within 20% or 40% Glycerol
different blood services, the shelf life of different components o cryoprotectant
varies considerably. Shelf-life specifications must comply with local Rapid freezing and stored in a freezer
standards. Transfusion of frozen cells must be followed by deglycerolization
Cryoprotectant us systematically replaced with decreasing
For Red Cells concentrations of saline
Shelf life varies at +4°C ± 2°C according to anticoagulant/preservative Current Trend: FDA licenses frozen RBCs for a period of 10 years
and additive solution used. from the date of freezing
The requirement that determines shelf life is that at least 75% of red
cells transfused at the end of the proposed storage period must still be
in circulation 24 hours after transfusion.

#MLSWATER2022 Page 10 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
RBC REJUVENATION pH 6.2: current standard for maintaining satisfactory platelet
Process of enhancing and restoring ATP & 2,3-DPG levels viability
RBCs (liquid state) can be rejuvenated at outdate or up to 3 days Second generation storage containers
after outdate o Increased gas transport properties
o Ex. Rejuvesol
Incubation of RBC unit with 50 mL of the rejuvenating solution for 1 PLATELET STORAGE AND BACTERIAL CONTAMINATION
hour at 37°C Storage of platelets at 20-24°C may cause bacterial growth
Following Rejuvenation, the RBCs can be washed with Saline & o Room temperature storage and the presence of Oxygen can
Transfused within 24 hours encourage bacterial proliferation
o Sepsis due to contaminated platelets: most common infectious
PLATELET STORAGE complication of transfusion
o 10-40% of patients transfused with a bacterially contaminated
MAJOR CHALLENGE IN PLATELET STORAGE
platelet unit develop life-threatening sepsis
5-day shelf life (in the US)
Bacterial contamination at incubation of 22°
A varying degree of platelet activation/ aggregation
INDICATIONS FOR BLOOD COMPONENTS
Release of intracellular granules RED BLOOD CELL TRANSFUSION
Suppression in ATP &ADP levels Are used to treat hemorrhage and to improve oxygen delivery to
tissues.
CLINICAL USE OF PLATELET It should be based on the patient's clinical condition.
Treatment of bleeding associated with thrombocytopenia Indications for transfusion include:
Transfused to hematology-oncology thrombocytopenic patients to o Symptomatic Anemia (causing shortness of breath, dizziness,
prevent bleeding congestive heart failure, and decreased exercise tolerance)
Current Trend: Platelets are prepared as concentrates from whole o Acute Sickle Cell Crisis
blood & increasingly by apheresis o Acute Blood Loss of more than 30 percent of blood volume.
Fresh frozen plasma infusion can be used for reversal of
CURRENT CONDITION OF PLATELET PRESERVATION anticoagulant effects.

PLATELET CONCENTRATES
Prepared from whole blood and apheresis components
PLATELET TRANSFUSION
Indicated to prevent hemorrhage in patients with
Storage: 20-24°C with continuous agitation
thrombocytopenia or platelet function defects.
Expiration time (FDA): midnight of Day 5
Cryoprecipitate is used in cases of hypofibrinogenemia, which most
Should contain a minimum of 5.5 x 10^10 platelets in a volume of
often occurs in the setting of massive hemorrhage or consumptive
between 45 & 65 mL
coagulopathy.
Transfusion-related infections are less common than non-infectious
RATIONALE AND ADVANE CONCEPT FOR PRESERVATION complications.
Maintenance of pH o All noninfectious complications of transfusion are classified as
o Key parameter for retaining platelet viability in vivo when noninfectious serious hazards of transfusion.
platelets were stored at 20-24°C o Acute complications occur within minutes to 24 hours of the
transfusion
#MLSWATER2022 Page 11 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o Delayed complications may develop days, months, or even years Rule of thumb:
later. A unit apheresis platelet (or a pool of 6 platelet concentrates)
Platelet transfusion may be indicated to prevent hemorrhage in should achieve an increment of 30,000 to 50,000/μL in an
patients with thrombocytopenia or platelet function defects. average adult. Alternative way to determine whether a
Contraindications to platelet transfusion include thrombotic patient is refractory is by calculating the corrected count
thrombocytopenic purpura and heparin-induced increment or CCI.
thrombocytopenia.
o Should not be transfused in patients unless a life-threatening Corrected count increment (CCI) and percent platelet recovery
hemorrhage has occurred. (PPR) are measures of response to platelet transfusion that
o Transfusion of platelets in these conditions can result in further "correct" the count increment for blood volume and number of
thrombosis. platelets transfused.
o One unit of apheresis platelets should increase the platelet count Corrected count increment (CCI) is a measure of the expected
in adults by 30 to 60 × 103 per μL (30 to 60 × 109 per L). increase in platelets following a platelet transfusion.
o In neonates, transfusing 5 to 10 mL per kg of platelets should o The “count increment” refers to the increase in platelets
increase the platelet count by 50 to 100 × 103 per μL (50 to 100 × following a transfusion.
109 per L). o The “correction” is based on the patient’s size and the number of
o One apheresis platelet collection is equivalent to six pooled platelets transfused.
random donor platelet concentrates. o It is used because CCI is a more accurate measure of
refractoriness, as it adjusts for the number of platelets
DISEASES IMPORTANT IN BLOOD BANKING transfused and the patient's blood volume.
o Immunodeficiency o It can guide the decision to pursue platelets with improved
o Hypersensitivity compatibility (i.e., HLA-matched platelets).
o Monoclonal and polyclonal gammopathies
o Autoimmune disease FORMULA:
o Hemolytic Disease of Newborn (HDN)
Further discuss in Module 1- Lesson 4 CCI = (count increment, per μL) × body surface area (m2) / unit
content
IMPORTANCE OF PLATELET CORRECTED COUNT INCREMENT (CCI)
Refractory is defined as failure to achieve acceptable increase in Units (m2/μL) are usually omitted when reporting the result.
platelet count following platelet transfusion on at least two
occasions and no alternate cause for refractoriness such as fever, Example:
sepsis, DIC, bleeding, splenomegaly, or drug interaction (e.g. For an adult patient with a BSA of 2.0 m2 whose platelet count rose
amphotericin B). from 5 × 109/L to 25 × 109/L after a platelet transfusion containing
Platelet count must be measured within one hour after transfusion. 4.0 × 1011 platelets:
Response to platelet transfusion is typically measured within one CCI for above patient = (20 × 109/L × 2.0 m2) / 4.0 = 10,000
hour. However, a sample collected 10 minutes after transfusion
yields similar information and may be easier to obtain routinely. A
post-transfusion platelet count can alternatively be taken at 20
hours.

#MLSWATER2022 Page 12 of 13
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
APPENDIX

SUPPLEMENTARY NOTES:
1. Glycolysis Reaction:
https://www.youtube.com/watch?v=uWOURkrxpH4

2. Luebering-Rapaport Pathway:
https://www.youtube.com/watch?v=ZB2yaxi4zV0&feature=youtu.be

-END OF TRANSCRIPTION-
Sana’y mahaba din pasensya mo this sem. Kasi paubos na yung sakin.
Charr : ))

#MLSWATER2022 Page 13 of 13
MODULE 1: Introduction to Immunohematology (part 3)
MLS 18, February 23, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
o The pairing that occurs is specific: adenine is partnered with
Legend: guanine and thymine is partnered with cytosine. Through the
Transcription Bullet process of binding, the strands twist and form a double helix.
Notes/Module Packet Bullet The double helix DNA forms an actual unit of inheritance, a gene.
MODULE OUTLINE
I. Historical Background and the current trends in Transfusion Medicine GENES
II. Blood Preservation Genetics is the study of inheritance (transmission of characteristics)
from parent to offspring
III. Review of Basic Genetics
o It is based on the biochemical structure of chromatin
A. Molecular Genetics o Chromatin is composed of nucleic acids, structural proteins and
B. Population Genetics enzymes.
C. Cellular Genetics The gene is an area of DNA that controls a trait or characteristic.
IV. Review of Fundamentals of Immunology The product of the gene is usually a protein or RNA.
V. Review of Molecular Biology
CENTRAL DOGMA OF MOLECULAR BIOLOGY
LEARNING OUTCOMES First described by James Watson and Francis Crick
1. Explain Mendel’s laws of independent segregation and random Framework for understanding the transfer of sequence
assortment. information between Information-carrying Biopolymers in living
2. Correlate the concepts of dominance and recessive traits with organisms
examples of the inheritance of blood group antigens.
3. Describe the process of mitosis and meiosis. DNA
4. Differentiate some of the ways in which Genetics can be used in Dictate the synthesis of RNA.
modern transfusion laboratory. Produce new DNA by replication
Produce RNA by transcription

MOLECULAR GENETICS: BASIC GENETIC COMPONENTS RNA


DNA Dictate the synthesis of Proteins
Produces proteins by translation
Deoxyribonucleic acid, the main building block of genetic material
Composed of four building blocks: adenine, guanine, cytosine, and
PROTEINS
thymine. These building blocks are bases.
Dictate the function of the cell
o Each base attaches to a sugar molecule and a phosphate molecule.
These building blocks form strings. Following the “stringing” of a
The location of a specific gene on the chromosome is known as its
single strand, there is a “pairing” process where the partner for each
genetic locus.
building block attaches and forms a double strand.
The alternate gene forms for a specific locus are known as alleles.
#MLSWATER2022 Page 1 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o For example, eye color might be determined by allele: blue, brown, CODOMINANCE is the expression of two different genes that are
hazel, or green. inherited at the same loci on a pair of chromosomes.
A single allele for eye color would be inherited from each parent. This
simple pattern of inheritance of a single allele from each parent will be With rare exceptions, blood group systems are expressed as codominant
applicable for blood group antigen inheritance. characteristics. For example, when an individual inherits an A gene from
When multiple alleles exist at a single locus, this is known as one parent and a B gene from the other parent, the genotype is AB. The
polymorphism. expressed blood type, or phenotype, will be AB. In this case, neither the A
o Some systems are more polymorphic than others. nor B gene is expressed in a dominant manner.
o A trait that has ten possible alleles at a single locus will be more
polymorphic than a trait with four possible alleles. AMORPH are genes that do not code for the production of any
o Example of a polymorphic gene is the ABO blood group detectable product. These genes appear to be recessive.

Antithetical genes are a pair (or more than 2) of genes that are loated in When the amorphic gene is inherited in conjunction with an allele that
different alleles does produce a detectable product, the detectable product of that allele
o Opposite form of gene is expressed. This allele is not dominant over the amorph nor is the
amorph recessive to the expressed allele. A common example of an
POLYMORPHISM of a locus determines the likelihood that two individuals amorph is the gene that codes for the O blood group. When inherited in
will be found with an identical allelic composition. a homozygous state, two O genes, there is no detectable product.
Hundreds of genes comprise each chromosome.
Humans have 23 pairs of chromosomes.
o Each individual inherits one half of his or her chromosomes from HARDY-WEINBERG PRINCIPLE
each parent. Mathematical equation to predict allele distribution and frequencies in
o The pairs consist of 22 pairs of autosomal chromosomes and one a population
pair of sex chromosomes. Allows for the study of Mendellian inheritance in great detail
o With rare exception, genes that code for blood group antigens and o Predictions are not always accurate
the production of blood group antigens are found on the autosomal Formula:
chromosomes.
p+q=1
GENE EXPRESSION o p = percentage/ gene frequency for the dominant allele
With inherited physical traits, there are certain patterns of expression. o q = percentage/ gene frequency for the recessive allele
Gene expression of simple physical traits typically follows one of three Also expressed by
patterns.
A gene is dominant if it is always expressed even when found in p2+2pq+q2=1
combination with a second gene. In this case, the second gene is not o p2 = Homozygous dominant allele (AA)
expressed. o q2 = Homozygous recessive allele (aa)
The “silent” recessive gene can only be expressed if two identical genes o 2pq = Heterozygous allele (Aa)
are present.
A third level of gene expression is codominant.

#MLSWATER2022 Page 2 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
ZYGOSITY The steric arrangement of two genes may create a weakened
ZYGOSITY describes the similarity or dissimilarity of genes at an allelic expression of one of the gene products.
position on two homologous chromosomes. o This is a position effect or steric hindrance of that particular gene
o When the genes are identical, they are said to be homozygous. product.
Conversely, when the genes are different, they are said to be An example of gene interaction exists within the Rh blood group system.
heterozygous. o D and C are specific genes in this system.
o All genes in the Rh system are inherited on the same chromosome.
The concept of zygosity can be related to antigen strength. o When C and D genes are inherited trans to each other (C on one
o There are some gene products that exhibit dosage. chromosome and D on the opposite homologous chromosome), the
o When genes are inherited as homozygous (i.e. two identical alleles steric effect will weaken the expression of the D antigen.
coding for the same product), the individual is said to have a o When the genetic relationship is cis (D and C on the same
“double dose” of this product. The expression of the trait or homologous chromosome), there is no effect on the antigen
product is stronger when present in the homozygous state. expression. For each genetic characteristic, an individual receives a
o In comparison, an individual that inherits a heterozygous set of gene from each of his or her parents. The sum total of both genes is
genes (i.e. two different alleles coding for two different products) is known as the individual’s genotype.
said to have a “single dose” of each gene since only one gene for For example, the inheritance of a “blue” gene from the mother and a
each product has been inherited. In this case, the expression of each “blue” gene from the father produces a genotype of “blue/blue.” This
trait or product is weaker than when two identical genes are individual is homozygous for the “blue” allele.
inherited. An individual that inherits a P gene from his or her mother and a Q gene
o Dosage is exhibited with some blood group systems. Red cells that from his or her father has a genotype of P/Q and is heterozygous for the
are heterozygous for a specific antigen will demonstrate a weaker P/Q alleles.
reaction than the homozygous cells when they react with the
specific antisera. NOTE:
The frequency of each genotype is reflective of the degree of
This is an important concept in blood group testing. polymorphism within that system.
o Interactions may occur between two genes. Systems that display a greater degree of polymorphism (i.e. more
o Interactions are dependent on location of the inherited genes. The alleles at each locus) will have a lower frequency for each allele than
likelihood of an interaction occurring is determined by the those that are less polymorphic.
proximity of the genes on the autosomal chromosomes.

Cis- PHENOTYPE
Relationship of two genes that are inherited on the same The phenotype is a function of gene expression.
chromosomes o The product of a recessive gene will not be expressed in a
Gene expression is stronger compared to Trans genes phenotype.
Trans- The dominant gene will produce a detectable product, whether in the
Genes that are inherited on different/ seperate chromosomes. homozygous or heterozygous state.
If the alleles are codominant, both will be expressed in the phenotype.

#MLSWATER2022 Page 3 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o In a system where the alleles are codominant, an individual with a o The F2 generation has a ratio of three red-flowered plants to one
genotype of Z/Y will have a phenotype of ZY while an individual with white flowered plant.
the genotype Z/Z will have a phenotype of Z. o This is because the plants that have the R gene, either RR
homozygous or Rr heterozygous, will have red flowers because the
POPULATION GENETICS: MENDEL’S LAWS red gene is dominant.
Gregor Mendel is recognized as the father of genetics. o Only when the red gene is absent and the white gene occurs in
o The results of his genetic research can be directly applied to the duplicate, as in the rr homozygous white-flowered plant, will the
inheritance of blood group antigens. recessive white gene expression be visible as a phenotype.
o Mendel was an Austrian monk and mathematician who used sweet This illustrates Mendel’s first law, the law of independent segregation.
pea plants growing in a monastery garden to study physical traits in o Therefore, each gene is passed on to the next generation on its
organisms and how they are inherited. own.
He first described the law known as independent segregation which o Specifically, Mendel’s first law shows that alleles of genes have no
refers to the transmission of a trait between generations in a permanent effect on one another when present in the same plant
predictable fashion. but segregate unchanged by passing into different gametes.
He determined the physical traits to be due to factors he called
elementen (eventually known as genes) within the cell.
He studied the inheritance of several readily observable pea plant (a
good model organism) characteristics
o notably flower color, seed color, and seed shape—and based his
first law of inheritance, the law of independent or random
segregation, on these results.

MENDEL’S 1ST LAW: INDEPENDENT SEGREGATION


The first generation in the study, called the parental, pure, or P1
generation, consisted of all red or all white flowers that bred true for
many generations.
The plants were either homozygous for red flowers (RR, a dominant
trait; dominant traits are usually written with uppercase letters). or
homozygous for white flowers (rr, a recessive trait; recessive traits are
usually written with lowercase letters).
o When these plants were crossbred, the second generation, called
first-filial, or F1, had flowers that were all red.
o Thus the dominant trait was the only trait observed.
When plants from the F1 generation were crossbred to each other, the
second-filial, or F2, generation, of plants had flowers that were red and
white in the ratio of 3:1.
o All the plants from the F1 generation are heterozygous (or hybrid)
for flower color (Rr).
An intermediate situation can also occur when alleles exhibit partial
dominance.
#MLSWATER2022 Page 4 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o This is observed when the phenotype of a heterozygous organism is
a mixture of both homozygous phenotypes seen in the P1
generation.
o An example of this is plants with red and white flowers that have
offspring with pink flowers or flowers that have red and white
sections.
o It is important to remember that although the phenotype does not
show dominance or recessive traits, the F1 generation has the
heterozygous genotype of Rr. It is essential to understand how a
genotype can influence a phenotype, and using flower color is a
good basic model system to study this.

Unlike the flower color of many types of plants, most blood group genes
are inherited in a codominant manner.
o In codominance, both alleles are expressed, and their gene
products are seen at the phenotypic level. PEDIGREE CHARTS
o In this case, one gene is not dominant over its allele, and the protein A method for tracking family history and inheritance patterns is a
products of both genes are seen at the phenotypic level. pedigree chart.
o An example of this is the MNSs blood group system, in which a It is a visual representation of the parents and the possible genotypes
heterozygous MN individual would type as both M and N antigen and phenotypes for the offspring.
positive. This chart illustrates the inheritance patterns of all the family
o Blood group antigens are inherited in this fashion with only rare members and can be used for visualization of inherited traits,
variations. The outcome of this type of inheritance can be predicted including blood group systems.
by a Punnett Square. o The pedigree chart is useful since it is more detailed than the
Punnett Square.
PUNNETT SQUARES
Prediction of possible genotypes and phenotypes in an offspring can MENDEL’S 2ND LAW: INDEPENDENT ASSORTMENT
be performed using a Punnett Square. The second concept of Mendelian genetics/ Mendel’s second law
In order to use the Punnett Square as a predictive tool, the user must States that genes for different traits are inherited separately from
know the exact genotype or inferred genotype of both parents. each other.
Using this information, a Punnett Square can be set up to predict the o This allows for all possible combinations of genes to occur in
likelihood of the genotype and phenotype of the offspring. the offspring.
o Specifically, if a homozygote that is dominant for two
different characteristics is crossed with a homozygote that is
recessive for both characteristics, the F1 generation consists
of plants whose phenotype is the same as that of the
dominant parent.

#MLSWATER2022 Page 5 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o However, when the F1 generation is crossed in the F2 Linkage can be determined by examining the frequency of the antigen
generation, two general classes of offspring are found. or product in the general population.
One is the parental type; the other is a new phenotype called a A more complex example of linkage disequilibrium occurs within the
reciprocal type and represents plants with the dominant feature of HLA antigen system.
one plant and the recessive feature of another plant. o The HLA-A and HLA-B antigens are more closely linked than
o Recombinant types occur in both possible combinations. the M/N and S/s genes.
Mendel formulated this law by doing studies with different types of o An individual’s haplotype is the set of HLA antigens inherited
seeds produced by peas and noted that they can be colored green or from one parent.
yellow and textured smooth or wrinkled in any combination. o For example, the mother of an offspring may be typed as HLA-
Genes located on different chromosomes are inherited separately A3, A69; B7, B45. This mother may pass along to herprogeny
and expressed discreetly from one another. the haplotype A3, B7, or A69, B45 but neverA3, B45, or A69,
o In most cases, this applies to the blood group antigens. B7.
Even blood groups that are inherited on the same chromosome, such NOTE:
as Rh and Duffy (Chromosome #1), are inherited as separate entities. Aside from Mendel’s work other major areas of population genetics are
o One is not dependent on the other for inheritance or due to:
expression.
 The pioneering work of Linnaeus and Darwin
o Separate genes account for the inheritance of blood group
system antigens.  Hardy-Weinberg Principle
 Inheritance patterns
LINKED GENES
Genes are in very close proximity, that are inherited as a unit rather
than as separate entities. CONCEPTS OF DOMINANCE AND RECESSIVE TRAITS
Independent assortment does not occur with antigens that are A variation on incomplete dominance is wherein both alleles for the
linked. same characteristic are simultaneously expressed in the
For example, the two genes that code for M/N and S/s antigen pairs heterozygote.
are very close to one another. They are inherited as a “package” An example of codominance occurs in the ABO blood groups of
from each parent. humans. The A and B alleles are expressed in the form of A or B
Two genes are linked if the products appear with greater molecules present on the surface of red blood cells.
frequency than expected if inherited independently Homozygotes (IAIA and IBIB) express either the A or the B phenotype,
o This deviance from anticipated frequencies is termed linkage and heterozygotes (IAIB) express both phenotypes equally.
disequilibrium. o The IAIB individual has blood type AB.
In a self-cross between heterozygotes expressing a codominant trait,
the three possible offspring genotypes are phenotypically distinct.
LINKED GENES
o However, the 1:2:1 genotypic ratio characteristic of a
Set of genes inherited via one of the two parental gamates Mendelian monohybrid cross still applies.

#MLSWATER2022 Page 6 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
MULTIPLE ALLELES
Although individual humans (and all diploid organisms) can only have
two alleles for a given gene, multiple alleles may exist at the
population level, such that many combinations of two alleles are
observed.
o Mendel implied that only two alleles, one dominant and one
recessive, could exist for a given gene, which is an
oversimplification.
Note that when many alleles exist for the same gene, the convention is
to denote the most common phenotype or genotype in the natural
population as the wild type (often abbreviated “+”).
All other phenotypes or genotypes are considered variants (mutants) of
this typical form, meaning they deviate from the wild type.
o The variant may be recessive or dominant to the wild-type allele.
An example of multiple alleles is the ABO blood-type system in humans.
o In this case, there are three alleles circulating in the population. The
IA allele codes for A molecules on the red blood cells, the IB allele Notice that instead of three genotypes, there are six different
codes for B molecules on the surface of red blood cells, and the i genotypes when there are three alleles.
allele codes for no molecules on the red blood cells. o The number of possible phenotypes depends on the dominance
o In this case, the IA and IB alleles are codominant with each other relationships between the three alleles.
and are both dominant over the i allele.
o Although there are three alleles present in a population, each
individual only gets two of the alleles from their parents.
o This produces the genotypes and phenotypes shown in the table GENOME
below. It takes two gametes to make a fertilized egg with the correct (2N)
number of chromosomes in the nucleus of a cell.
DOSAGE EFFECT o Therefore, each parent contributes only half (1N) of the inherited
Excess unbound immunoglobulin that leads to Prozone effect genetic information, or genes, to each child.
An antibody gives a stronger reaction with RBC double-dosed for the In order to be completely healthy, each child must have the correct
target antigen number of genes and chromosomes (2N), without major mutations
affecting necessary biochemical systems.
A homozygous recessive trait may be expressed more strongly than a
o At the smallest level, genes are composed of discrete units of DNA
heterozygous trait
arranged in a linear fashion, similar to a strand of pearls, with
Affected by inheritance of genotype
structural proteins wrapped around the DNA at specific intervals to
pack it into tightly wound bundles.
PROZONE EFFECT
effectiveness of antibodies to form immune complexes is sometimes
impaired when concentrations of an antibody or an antigen are very
high

#MLSWATER2022 Page 7 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
CHROMOSOMES are DNA that is organized at a higher level CELLULAR GENETICS: CELL DIVISION
o Each chromosome being one incredibly long strand of duplex In eukaryotic cells such as human cells, the cell cycle is divided into four
(double-stranded) DNA. distinct stages and is represented by a clock or circular scheme
A GENE is a section, often very large, of DNA along the chromosome. It indicating that it can repeat itself or can be stopped at any one point in
controls a trait or characteristic the cycle.
o The specific sequence of nucleotides and the location on the
chromosome determines a gene. G0 / resting stage The first step, the state of cells not actively
o In addition, each gene has specific and general sequences that dividing.
occur upstream (before the start site) and downstream (after the G1 pre-replication stage
termination signals) that contribute to how the gene functions. S Phase The step at which DNA is synthesized
RNA is the product of a gene, that codes for proteins G2 Postreplication stage
A LOCUS (plural = loci) is the specific location of a gene on a M phase mitosis occurs
chromosome
ALLELES are specific locus formed by an alternate gene. They one or Chromosomes are in the interphase stage of mitosis in the span from
several different forms of the same gene at each locus G0 to the end of the G2 phase.
GENOTYPE is the sequence of DNA that is inherited. Cells that are completely mature and no longer need to divide to
PHENOTYPE is anything that is physically manifested; the ratio of increase their numbers, such as nerve cells, can remain in the G0 stage
muscle fibers; the level of hormones produced; and such obvious traits for a very long time.
as eye, skin, and hair color. It is a hallmark of cancer cells, such as the transformed cells seen in the
various leukemias and solid tumors, that they can go through the stages
KEEP IN MIND that more than one gene can affect a particular trait (part of cell division much faster than non-transformed, normal cells and
of a phenotype), such as the height of an individual; all relevant genes can therefore outgrow them.
be considered as part of the genotype for that trait. Depending on the o In this way, they take up the bulk of nutrients needed by the
alleles inherited, an organism can be either homozygous or heterozygous nontransformed cells and crowd them out of existence and
for a specific trait. The presence of two identical alleles results in a potentially overgrow the adult organism in which they occur.
homozygous genotype (i.e., AA), and the phenotype is group A blood. On
the other hand, the inheritance of different alleles from each parent gives MITOSIS
a heterozygous genotype.
The process by which cells divide to create identical daughter cells is
called mitosis
Another important concept is that of the “silent” gene, or amorph, and During cell division, the chromosomes are reproduced in such a way
the term hemizygous. that all daughter cells are genetically identical to the parent cell.
o Without maintaining the same number and type of chromosomes,
An AMORPH is a gene that does not produce any obvious, easily the daughter cells would not be viable.
detectable traits and is seen only at the phenotypic level when the The chromosomes are duplicated, and one of each pair is passed to the
individual is homozygous for the trait. daughter cells.
During the process of mitosis, quantitatively and qualitatively identical
HEMIZYGOUS refers to the condition when one chromosome has a copy DNA is delivered to daughter cells formed by cell division.
of the gene and the other chromosome has that gene deleted or absent. The complex process of mitosis is usually divided into a series of stages,
characterized by the appearance and movement of the chromosomes.
#MLSWATER2022 Page 8 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
DNA is in the form of chromatin and is dispersed MEIOSIS I The first stages of meiosis are nearly identical to
throughout the nucleus. This is the stage of the DNA those in mitosis, in which chromatin is
INTERPHASE
when cells are not actively dividing. New DNA is condensed, homologous chromosomes are
synthesized by a process called replication. paired in prophase, and chromosomes are
The chromatin condenses to form chromosomes. In aligned along the center of the cell.
PROPHASE
prophase, the nuclear envelope starts to break down. However, there is no centromere division, and at
The chromosomes are lined up along the middle of anaphase and telophase, the cell divides and
the nucleus and paired with the corresponding enters interphase once again, in which there is
METAPHASE
chromosome. In this stage, chromosome preparations no replication of DNA.
are made for chromosome analysis in cytogenetics.
The cellular spindle apparatus is formed and the PROPHASE II Chromosomes are condensed
chromosomes are pulled to opposite ends of the cell. METAPHASE II centromeres divide and chromosomes line up along
ANAPHASE
The cell becomes pinched in the middle, and cell the center again
division starts to take place ANAPHASE II Chromosomes are pulled to opposite ends of the cell
the cell is pulled apart, division is complete, and TELOPHASE II The two cells divide, giving rise to four 1N daughter
the chromosomes and cytoplasm are separated cells.
TELOPHASE
into two new daughter cells.
In addition, during meiosis, crossing over and recombination can
happen between maternal- and paternal-derived chromosomes.
MEIOSIS o This allows for the creation of new DNA sequences that are
A different process is used to produce the gametes or sex cells. different from the parent strains. Combined with random
The process results in four unique, rather than two identical, daughter segregation, it is possible to have very large numbers of new DNA
cells. sequences.
o The uniqueness of the daughter cells generated with meiosis allows o In humans with 23 pairs of chromosomes, the total possible number
for great genetic diversity in organisms and controls the number of is several million.
chromosomes within dividing cells.
If cells with 2N chromosomes were paired, the resulting daughter cells
would have 4N chromosomes, which would not be viable. Therefore,
gametes carry a haploid number of chromosomes, 1N, so that when
they combine, the resulting cell has a 2N configuration.

Meiosis only occurs in the germinal tissues and is important for


reproduction.
o Without the complicated process of meiosis, there would be no
change from generation to generation, and evolution would not
occur or happen too slowly for organisms to adapt to environmental
changes.

#MLSWATER2022 Page 9 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
PATTERNS OF INHERITANCE
AUTOSOMAL DOMINANT
Genes expressed with equal frequency in males and females
Non-sex chromosome
One parent can transmit a trait to a son or daughter

SEX-LINKED DOMINANT
Trait carried on the X chromosome
Father to daughter transmission only
o No father to son transmission

SEX-LINKED RECESSIVE
Trait carried on the X chromosome
Carrier mother to son transmission
Traits are exhibited most commonly in males
o Example: Hemophilia A

-END OF TRANSCRIPTION-
….for now (0_0)

#MLSWATER2022 Page 10 of 10
MODULE 1: Introduction to Immunohematology (part 4)
MLS 18, February 23, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
Cellular Immunity
Legend: Mediated by various IS cells, such as macrophage, T cells, and
Transcription Bullet dendritic cells.
Notes/Module Packet Bullet Lymphokines- are other effector molecules that play critical roles in
MODULE OUTLINE the cellular system by activating and deactivating different cells,
I. Historical Background and the current trends in Transfusion Medicine which allows cells to communicate throughout the host body. It
II. Blood Preservation includes cytokines and chemokines.
III. Review of Basic Genetics
Humoral Immunity
IV. Review of Fundamentals of Immunology
Consists of the fluid parts of the IS, such as antibodies and
V. Review of Molecular Biology complement components found in plasma, saliva, and other
secretions.
LEARNING OUTCOMES Antibody- also known as immunoglobulins (immune because of
At the end of this module, the student shall be able to: their function and globulin because they are a type of globular
1. Outline the cellular and humoral components of the immune soluble protein). Its function is to bind to foreign molecules called
system. antigens.
2. Describe the characteristics of immunoglobulins in relation to its o Antigen-antibody reactions are specific. Only one antibody
significance in transfusion medicine. reacts with one antigen or one part (an epitope/antigenic
3. List the methods used in the blood bank to detect antibodies and determinant) of a complex antigen.
complement bound to red blood cells.
4. Identify the various factors that affect agglutination reactions. Innate Immunity
5. Describe some of the common diseases that can affect blood bank
It is the immediate line of immune defense.
testing.
First line of defense, not specific.
It doesn’t need modification to function to the same antigen
WHAT IS IMMUNITY? Physical barriers that includes are:
Refers to the process by which a host organism protects itself from o skin
attacks by external and internal agents. o mucous membrane
Confers protection from non-self and abnormal self-elements which o cilia lining
are controlled at different levels. o cough mechanism
The response and elimination of organisms and unwanted cells is
accomplished through cellular and/or humoral mechanisms. Biochemical barriers
o bactericidal enzyme (lysozymes, RNase, fatty acid, sweat,
digestive enzyme, stomach acid, and vaginal low ph)

#MLSWATER2022 Page 1 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
Acquired/ Adaptive Immunity Characteristics of Immunoglobulins
Acquired refers to the fact that the immunity is acquired via specific A complex protein produced by plasma cells, with specificity to
contact with a pathogen or aberrant cell. antigens that stimulate their production.
Adaptive refers to the ability to adapt to and destroy new complex 20% Immunoglobulins present to an individual
pathogens, although it must first react to them through complex Antibodies bind antigen, fix complement, facilitate phagocytosis and
recognitions processes. neutralize toxic substances in the circulation.
They are classified according to the molecular structure of their
heavy chains. Five classifications:
CELLS AND ORGANS OF THE IMMUNE SYSTEM
Different types of immune system cells can be distinguished by the IgG Most concentrated in serum (80%) of total
membrane markers they possess known as clusters of differentiation (gamma heavy chain) serum Ig.
(CD) markers and are detected by immunotyping methods. secreted by the plasma in the blood
acts in long term immune system
Types of Cells: ability to cross the placenta
1. Lymphocytes (B cell/T cell) IgA Is next with 13% (although majority it is
2. Plasma cell (alpha heavy chain) major found in body secretions)
3. Natural Killer cells found in mucous, saliva, tears
present in secretions
B cells IgM 6%
Antibodies are secreted by mature B cells called plasma cells and bind (mu heavy chain) responsible for the early stages of
to antigens in specific manner. immunity
When the receptor on the B cell reacts with a specific antigen and 1gD 1%
recognizes it, the B cell is activated to divide. The cells produced from (delta heavy chain) receptor in B cells
this rapid division mature into plasma cells and memory B cells. triggers or activates basophils or mast
Memory B cells have antibody on their surfaces that is of the same cells
conformation as that of the B cell from which they were derived. found in the membrane of immature B
cell
helps in the activation of plasma cell
T cells
IgE Least common, <1%
(epsilon heavy chain) present against allergic reaction
They require help in the form of cell membrane proteins known as
protect against parasitic worms
major histocompability complex (MHC) molecules
MHC 2 classes
Immunoglobulin Structure
o Class 1: found in all nucleated cells
o Class 2: found in APC (Antigen Presenting Cell) Basic Ig structural unit is composed of four polypeptide chains; two
The MHC genes determine the human leukocyte antigens (HLA) identical light chains (MW=approximately 22,500 daltons) and two
present on leukocytes and other cells; they have been known for identical heavy chains (MW= app. 50,000 to 75,000 daltons).
many years to cause rejection of tissue grafts. Covalent disulfide bonding holds the light and heavy chains
T cell mediated immunity involve response against fungal and viral together. The covalent disulfide linkages in Ig molecules provide
infection, intracellular parasite, tissue grafts, and tumor.
#MLSWATER2022 Page 2 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
greater structural strength than hydrogen bonding and van der produced in response to commonly occurring antigens like intestinal
Waals forces. However, they limit the flexibility of the Ig molecule. flora and pollen grains.
The heavy chains are also interconnected by disulfide linkages in o Other blood groups such as Lewis, Ii, P and MNS may also
the hinge region of the molecule. produce IgM antibodies, which usually react best at ambient
Two types of light chains: kappa and lambda (both are present in temperature (22⁰C to 24⁰C)
all classes of immunoglobulins, regardless of the heavy chain The primary testing problem encountered with IgM antibodies is
classification). that they can interfere with the detection of clinically significant IgG
Ig molecules are protein and therefore have two terminal regions: antibodies by masking their reactivity. Unlike IgG, IgM exist both
the amino (-NH2) terminal and the carboxyl (-COOH) terminal. monomeric and polymeric forms (as pentamers) containing a J
Enzyme papain splits the antibody molecule at the hinge to give (joining) chain.
three fragments: one crystallizable Fc fragment and two antigen The pentameric form can be dissociated through cleavage of
binding fragments, Fab. covalent bonds interconnecting the monomeric subunits and the J
Domains of the immunoglobulins are the regions of the light and chain by chemical treatment with sulfhydryl reducing reagents such
heavy chains that are folded into compact globular loop structures. as ß-2-mercaptoethanol (2-ME) or dithiothreitol (DTT). These
reagents can distinguish a mixture of IgM and IgG antibodies
because only IgM is removed by the use of such compounds;
therefore, the removal allows unexpected IgG to be detected.

IgG
IgG antibodies are significant in transfusion medicine because they
are the class of immunoglobulins that are made in response to
transfusion with non-self and therefore are incompatible RBCs and
other blood products.
IgG antibodies are important in hemolytic disease of the newborn
(HDN) because antibodies can be formed in response against
alloantigens on fetal RBCs that enter the mother’s circulation,
usually during delivery.
IgG has the greatest number of subclasses: IgG1, IgG2 , IgG3 , and
IgG4 , and all four are easily separated by electrophoresis.
Immunoglobulins Significant for Blood Bank o The small differences in the chemical structure within the
All immunoglobulins can be significant for transfusion medicine; constant regions of the gamma heavy chains designate the
however, IgG, IgM and IgA have the most significance. various subclasses, and the number of disulfide bonds
Most clinically significant antibodies that react at body between the two heavy chains in the hinge region of the
temperature (37⁰C) are IgG isotype and are capable of destroying molecule constitutes one of the main differences between
transfused antigen-positive RBCs, causing anemia and transfusion subclasses.
reactions of various severities. Functional differences between the subclasses include the ability to
IgM fix complement and cross the placenta.
IgM antibodies are most commonly encountered as naturally
occurring antibodies in the ABO system and are believed to be

#MLSWATER2022 Page 3 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
IgA IgD
Like IgM, IgA exists in two main forms, a monomer and polymer IgD, present as less than 1 percent of serum immunoglobulins,
form, as dimers or trimers composed of two or three identical appears to have functions that deal primarily with maturation of B
monomers, respectively, joined by a J chain. cells into plasma cells. IgD is usually found bound to the membrane
IgA is located in different parts of the IS depending on subclass. of immature B cells. Therefore, IgD may be necessary for regulatory
o Serum IgA is found in both monomeric and polymeric forms; roles during B-cell differentiation and antibody production but is
Serum IgA can cause problems when transfused in probably the least significant for blood banking.
plasma product to patient having deficient IgA can cause
fatal anaphylaxis and can induce IgG rbc hemolysis CHARACTERISTICS IgG1 IgG2 IgG3 IgG4
o Secretory IgA is usually found in the mucosal tissues of the Proportion of 65-70 23-28 4-7 3-4
body. total serum IgG
• Its polymer form acquires a glycoprotein secretory (%)
component as it passes through epithelial cell walls of Complement ++ + +++ 0
mucosal tissues and appears in nearly all body fluids, fixation (classic
including saliva, tears, bronchial secretions, prostatic pathway)
fluid, vaginal secretions, and the mucous secretions of Binding to +++ ++ +++ +
the small intestine. macrophage FC
Also, anti-IgA antibodies can cause severe problems if transfused in receptors
plasma products to patients who are deficient in IgA, as potentially
Ability to cross + + + +
fatal anaphylaxis can result. Another reason for the importance of
placenta
IgA is that IgA can increase the effect of IgG-induced RBC hemolysis.
Dominant antibody activities:
Anti-Rh ++ 0 + +
IgE Anti-factor VII 0 0 0 +
IgE is normally found only in monomeric form in trace
Anti-dextran 0 + 0 0
concentrations in serum, about 0.004% of total immunoglobulins,
Anti-Kell + 0 0 0
and is important in allergic reactions.
Anti-Duffy + 0 0 0
The Fc portion of the IgE molecule attaches to basophils and mast
cells and facilitates histamine release when an allergen binds to the Anti-platelet 0 0 + 0
Fab portion of the molecule and cross- links with a second molecule Biological half-life 21 21 7-8 21
on the cell surface. (days)
Histamine is critical for bringing about an allergic reaction.
o Although hemolytic transfusion reactions are not caused by Immunoglobulin variations
IgE, urticaria may occur because of the presence of IgE Isotype (class variation)
antibodies. Refers to variants present in all members of a species, including the
o Because IgE causes transfusion reactions by release of different heavy and light chains and the different subclasses.
histamines, patients who have several allergic reactions to
blood products can be pretreated with antihistamines to
Allotypic
counteract the response when receiving blood products.
Present primarily in the constant region not all variants occur in all
members of a species.
#MLSWATER2022 Page 4 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
Idiopathic Lectin Pathway
which determines the antigen-binding specificity regions and is The lectin pathway is activated by attachment of plasma mannose-
specific for each antibody molecule. binding lectin(MBL) to microbes. MBL in turn activates proteins of
the classical pathway.
Immunoglobulin Fc Receptors
Macrophages and monocytes have receptors for the attachment Characteristics of Antigens
and can bind CH3 domain of the Fc portion. The immune response is initiated by the presentation of an antigen
Only the IgG1 and IgG3 subclasses are capable of attachment to (can initiate formation of and react with an antibody) or
phagocytic receptors. This is one way that incompatible RBCs coated immunogen (can initiate an immune response) to the IS and the IS
with IgG antibody are removed by phagocytosis. determining that the antigen is non-self.
The other phagocytic cells with Fc receptors include neutrophils, NK The immune reaction to any immunogen, including antigens, is
cells and mature B cells. determined by the host response as well as by several biochemical
and physical characteristics of the immunogen.
Complement system Properties such as size, complexity, conformation, charge,
Is a complex group of over 20 circulating and cell membrane accessibility, solubility, digestibility, and biochemical composition
proteins that have a multitude of functions within the immune influence the amount and type of immune response.
response Molecules that are too small cannot stimulate antibody production.
Primary roles include direct lysis of cells, bacteria, and enveloped Immunogens having a molecular weight (MW) less than 10,000
viruses as well as assisting with opsonization to facilitate daltons (D), for example, are called haptens and usually do not elicit
phagocytosis. an immune response on their own. A hapten coupled with a carrier
Another role is production of peptide fragment split products, which protein having a MW greater than 10,000 D, however, can produce
play roles in inflammatory responses such as increased vascular a reaction.
permeability, smooth muscle contraction, chemotaxis, migration The biochemical composition of the stimulus plays a role in immune
and adherence. stimulation.
The complement proteins are activated in a cascade of events Remember that RBC antigens are very diverse in structure and
through three main pathways: the classical, alternative and lectin composition and may be proteins (such as the Rh, M, and N blood
pathways. group substances) or glycolipids (such as the ABH, Lewis, Ii, and P
The three pathways converge at the activation of the complement blood group substances).
C3: Human leukocyte antigens (HLAs) are glycoproteins. Because of
these differences in structure, conformation, and molecular nature,
not all blood group substances are equally immunogenic in-vivo.
Classical Pathway
The classical pathway is activated by binding of an antigen with an
Characteristics of Blood Group
IgM, IgG1 or IgG3 antibody.
Polyclonal and Monoclonal Antibodies
Alternative Pathways Polyclonal or serum antibodies and are produced in response to a
Alternative pathway is activated by high molecular weight single antigen with more than one epitope.
molecules with repeating units found on the surfaces of target cells. Monoclonal antibodies- are produced by isolating individual B cells
from a polyclonal population and propagating them in cell culture
with hybridoma technology
#MLSWATER2022 Page 5 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
The supernatant from the cell culture contains antibody from a single same sample. These antibodies may be able to hemolyze,
type of B cell, clonally expanded, and therefore with the same agglutinate, or sensitize RBCs.
variable region and having a single epitope specificity. Some antibodies require special reagents to enhance their reactivity
This results in a monoclonal antibody suspension. Monoclonal and detection. Due to the enormous polymorphism of the human
antibodies are preferred in testing because they are highly specific, population, a diversity of RBC antigens exists, requiring a variety of
well characterized, and uniformly reactive. Most reagents used today standardized immunologic techniques and reagents for their
are monoclonal in nature. detection and identification.

Naturally Occurring and Immune Antibodies Alloantibodies and Autoantibodies


RBC antibodies are considered naturally occurring when they are Alloantibodies
found in the serum of individuals who have never been previously Alloantibodies are produced after exposure to genetically different,
exposed to RBC antigens by transfusion, injection, or pregnancy. or non-self, antigens of the same species, such as a different RBC
These antibodies are probably produced in response to substances antigen after transfusion.
in the environment that are highly similar to RBC antigens such as Transfused components may elicit the formation of alloantibodies
pollen grains and parts of bacteria membranes. against antigens (red cell, white cell and platelets) not present in the
Most naturally occurring antibodies are IgM cold agglutinins, which recipient.
react best at room temperature or lower; activate complement, and
when active at 37⁰C may be hemolytic. In blood banking, the Autoantibodies
common naturally occurring antibodies react with antigens of the Autoantibodies are produced in response to self-antigens. They can
ABH, Hh, Ii, Lewis, MN, and P blood group systems. cause reactions in the recipient if they have a specificity that is
RBC antibodies are considered immune when found in the serum of common to the transfused blood.
individuals who have been transfused or pregnant. These antigens Some autoantibodies do not have a detectable specificity and are
are not generally found in nature, and their molecular makeup is referred to as pan- or polyagglutinins.
unique to human RBCs. Autoantibodies can react at different temperatures, and cold or
Most immune RBC antibodies are IgG antibodies that react best at warm autoantibodies may both be present.
37⁰C and require the use of antihuman globulin sera (Coombs sera)
for detection. The most common immune antibodies encountered Characteristics of Antigen-Antibody Reactions
in testing include those that react with the Rh, Kell, Duffy, Kidd, and The antigen-binding site of the antibody molecule is uniquely
Ss blood group systems. designed to recognize a corresponding antigen; this antibody amino
acid sequence cannot be changed without altering its specificity.
Unexpected Antibodies The extent of the reciprocal relationship, also called the fit between
All other antibodies directed against RBC antigens are considered the antigen and its binding site on the antibody, is often referred to
unexpected and must be detected and identified before blood can as a lock and key mechanism.
be safely transfused, even if antibodies react at room temperature
or only with Coombs sera.
Also, autoreactive antibodies must be investigated. The reactivity of
unexpected antibodies is highly varied and unpredictable as they
may be either isotype IgM or IgG; rarely, both may be present in the

#MLSWATER2022 Page 6 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
Detection of RBC Antigen-Antibody Reactions Agglutination inhibition
Various factors influence detection of RBC antigen-antibody A method in which a positive reaction is the opposite of what is
reactions. These include having a correct sample (a sample that is normally observed in agglutination. Agglutination is inhibited when
stored under the right conditions) and the proper reagents an antigen-antibody reaction has previously occurred in a test system
performing the correct test and understanding how the test should and prevents agglutination.
be done. The antigen and antibody cannot bind because another substrate has
been added to the reaction mixture and blocks the formation of
Traditional Laboratory Methods antigen-antibody agglutinates
Commonly used techniques including hemagglutination (a special
type of agglutination), precipitation, agglutination inhibition, and RIA, ELISA and IF Techniques
hemolysis. Other techniques such as radioimmunoassay (RIA), radioimmunoassay (RIA), enzyme-linked immunosorbent assay
enzyme-linked immunosorbent assay (ELISA) or enzyme (ELISA) or enzyme immunoassay (EIA), and immunofluorescence (IF)
immunoassay (EIA), and immunofluorescence (IF), which quantifies techniques are immunologic methods based on quantization of
antigen or antibody with the use of a radioisotope, enzyme, or antigen or antibody by the use of a radioisotope, enzyme, or
fluorescent label, respectively, may be used in automated or fluorescent label, respectively.
semiautomated blood banking instrumentation.
Factors That Influence Agglutination Reactions
Hemagglutination Typical of most biochemical reaction systems, agglutination reactions
Are methods for the analysis of blood group antigen-antibody are influenced by the concentration of the reactants (antigen and
responses and typing for ABO, Rh, and other blood group antigens is antibody) as well as by factors such as pH, temperature, and ionic
accomplished by red cell agglutination reactions. strength. The surface charge, antibody isotype, RBC antigen dosage,
o Agglutination is a straightforward process and can be shown to and the use of various enhancement media, antihuman globulin
develop in two stages. In the first stage, called sensitization, reagents, and enzymes are all important in antigen-antibody
antigen binding to the antibody occurs. reactions.
o Epitopes on the surfaces of RBC membranes combine with the Centrifugation
antigen combining sites (Fab region) on the variable regions of It decreases reaction time by increasing the gravitational forces on
the immunoglobulin heavy and light. the reactants as well as bringing reactants closer together. Under the
o Antigen and antibody are held together by various non-covalent right centrifugation conditions, sensitized RBCs overcome their
bonds, and no visible agglutination is seen at this stage. In the natural repulsive effect (zeta potential) for each other and
second stage, a lattice-type structure composed of multiple agglutinate more efficiently
antigen-antibody bridges between RBC antigens and antibodies
is formed. A network of these bridges forms, and visible Antigen-Antibody Ratio
agglutination is present during this stage. Antigen and antibody have optimal concentrations; in ideal reactive
conditions, an equivalent amount of antigen and antibody binds. Any
Precipitation reaction deviation from this decreases the efficiency of the reaction and a loss
The development of an insoluble antigen-antibody complex, resulting of the zone of equivalence between antigen and antibody ratio that
from the mixing of equivalent amounts of soluble antigen and is necessary for agglutination reactions to occur.
antibody.

#MLSWATER2022 Page 7 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o An excess of unbound immunoglobulin leads to a prozone Factors that influence Agglutination Reactions: ENHANCEMENT MEDIA
effect, and a surplus of antigen (antigen-binding sites) leads
Protein Media
to a postzone effect.
Colloidal substances, or colloids are a type of clear solution that
o Another reason antigen amount may be altered is due to
contains particles permanently suspended in solution. Colloidal
weak expression of antigen on RBCs (dosage effect). Weak
particles are usually large moieties like proteins as compared with
expression occurs as a result of the inheritance of genotypes
the more familiar crystalloids, which usually have small, highly
that give rise to heterozygous expression of RBC antigens
soluble molecules, such as glucose, that are easily dialyzed.
and resultant weaker phenotypes.
The colloidal solutes can be charged or neutral and go into solution
because of their microscopic size. Colloids include albumin,
Effect of pH polyethylene glycol (PEG), polybrene, polyvinylpyrrolidone (PVP),
The ideal pH of a test system for antigen-antibody reactions is a and protamine. These substances work by increasing the dielectric
range between 6.5 and 7.5, which is similar to the pH of normal constant (a measure of electrical conductivity), which then reduces
plasma or serum. Exceptions include some anti-M and some Pr(Sp1) the zeta potential of the RBC.
group antibodies that show stronger reactivity below pH 6.5.
LISS Media
Temperature Low Ionic Strength Solution (LISS) Media (LISS), or low salt media,
Different isotypes of antibodies may exhibit optimal reactivity at generally contain 0.2 percent sodium chloride.
different temperatures. IgM antibodies usually react optimally at They decrease the ionic strength of a reaction medium, which
ambient temperatures or below 22⁰C, whereas IgG antibodies usually reduces the zeta potential and therefore allows antibodies to react
require 37⁰C. more efficiently with RBC membrane antigens.
LISS media are often used because they result in an increased rate of
Ig type antibody uptake during sensitization and a decreased reaction
Examples of IgM antibodies that have importance in blood banking incubation time (from 30 to 60 minutes to 5 to 15 minutes as
include those against the ABH, Ii, MN, Lewis (Lea , Leb ), Lutheran compared with protein potentiators such as albumin).
(Lua ), and P blood group antigens. However, they can result in false-positive reactions and may cause
Important IgG antibodies are those directed against Ss, Kell (Kk, Jsa , wasted time if reactions have to be repeated with albumin.
Jsb , Kpa , Kpb ), Rh (DCEce), Lutheran (Lub ), Duffy (Fya , Fyb ), and
Kidd (Jka , Jkb ) antigen. Polyethylene Glycol (PEG)
Polyethylene Glycol (PEG) and Polybrene are macromolecule
Enhancement media additives used with LISS to bring sensitized RBCs closer to each other
Agglutination reactions for IgM antibodies and their corresponding to facilitate antibody cross-linking and agglutination reactions. T
RBC antigens are easily accomplished in saline medium as these they are often used in place of albumin and have some advantages
antibodies usually do not need enhancement or modifications to and possible drawbacks. Polybrene can detect ABO incompatibility as
react strongly with antigens. well as clinically significant IgG alloantibodies, whereas PEG produces
Many of the commercially available enhancement media accomplish very specific reactions with reduction in false positive or nonspecific
this by reducing the zeta potential of RBC membranes. The net reactions.
negative charge surrounding RBCs (and most other human cells) in a PEG is considered to be more effective than albumin, LISS, or
cationic media is part of the force that repels RBCs from each other polybrene for detection of weak antibodies.
and is due to sialic acid molecules on the surface of RBCs.
#MLSWATER2022 Page 8 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
Proteolytic Enzymes allowing the Fab portions more flexibility in facilitating agglutination
Are protein molecules that function by altering reaction conditions reactions.
and bring about changes in other molecules without being changed
themselves. New and Nontraditional Laboratory Methods
Enzymes used in the detection and identification of blood group Flow Cytometry
antibodies include ficin (isolated from fig plants), papain (from Solid-Phase Adherence
papaya), trypsin (from pig stomach), and bromelin (from Gel Test
pineapple). RBC Affinity Column Test
The use of enzymes provides enhanced antibody reactivity to Rh,
Kidd, P1, Lewis, and I antigens and destroys or decreases reactivity to
Fya , Fyb, M, N, and S antigens DISEASES IMPORTANT IN BLOOD BANK SEROLOGIC TESTING
Immunodeficiency diseases
Antihuman Globulin (AHG) Reagents Can result from various defects in the IS at many different levels of
When RBCs become coated with antibody or complement or both immune function and may be congenital or acquired. They can
but do not agglutinate in regular testing, special reagents are needed result from defects in either innate or adaptive immunity or both;
to produce agglutination. the cause is often not known.
The direct antihuman globulin (AHG) test is designed to determine if
RBCs are coated with antibody or complement or both. Hypersensitivity (or allergy)
o Polyspecific AHG can determine if RBCs have been sensitized
An inflammatory response to a foreign antigen and can be cell- or
with IgG antibody or complement (components C3b or C3d) or
antibody-mediated or both. There are four different types of
both.
hypersensitive reactions, and the symptoms and treatment required
o Monospecific AHG reagents react only with RBCs sensitized with
for each are different. All four types can be caused by blood product
IgG or complement.
transfusions and may be the first sign of a transfusion reaction.

Chemical Reduction of IgG and IgM Molecules


Type I Reactions
The reagents generally act on covalent sulfhydryl bonds and facilitate
Also called anaphylaxis or immediate hypersensitivity, involves
antibody identification by removal of either IgG or IgM antibodies.\
histamine release by mast cells or basophils with surface IgE
Dithiothreitol (DTT) and ß-2-mercaptoethanol (2-ME) are thiol
antibody.
reducing agents that break the disulfide bonds of the J (joining) chain
It can occur in IgA-deficient individuals who receive plasma products
of the IgM molecule but leave the IgG molecule intact
containing IgA. Urticarial reactions (skin rashes) may also result
Another reagent, ZZAP, which consists of a thiol reagent plus a
from transfusion of certain food allergens or drugs in plasma
proteolytic enzyme, causes the dissociation of IgG molecules from
products.
the surface of sensitized RBCs and alters the surface antigens of the
RBC. Type II Reactions
Chemical reduction of the disulfide bond of the IgG molecule is also Reaction can involve IgG or IgM antibody with complement,
used to produce chemically modified reagents that react with RBCs in phagocytes, and proteolytic enzymes.
saline. Sulfhydryl compounds reduce the strong but less flexible HDN or transfusion reactions caused by blood group antibodies as
covalent disulfide bonds in the hinge region of the IgG molecule, well as autoimmune hemolytic reactions are all type II reactions.

#MLSWATER2022 Page 9 of 10
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
Type III Reactions Hemolytic Disease of the Newborn
Involve phagocytes and IgG and IgM and complement. Type III (HDN) can result when the maternal IS produces an antibody
reactions result in tissue damage from the formation of immune directed at an antigen present on fetal cells but absent from
complexes of antigen-antibody aggregates, complement, and maternal cells.
phagocytes and are therefore very serious. The mother is exposed to fetal RBCs as a result of feto-maternal
Penicillin and other drug induced antibodies can lead to hemolytic transfer of cells during pregnancy or childbirth.
reactions through type III hypersensitivity. o Maternal memory cells can cause a stronger response during a
second pregnancy if the fetus is positive for the sensitizing
Type IV Reactions antigens. IgG1, IgG3, and IgG4 are capable of crossing the
The type IV reaction involves only T cell–mediated responses and placenta and attaching to fetal RBCs whereas IgG2 and IgM are
their cytokines and can be fatal if untreated. The most important not.
type IV reaction is graft versus-host, of which there is also more o Severe HDN is most often associated with IgG1 antibodies and
than one type. may require exchange transfusion

Monoclonal and Polyclonal Gammopathies -END OF TRANSCRIPTION-


Plasma cell neoplasms result in proliferation of abnormal
immunoglobulin (or gamma globulin) from either a single B cell clone
(monoclonal gammopathies) or multiple clones (in polyclonal
gammopathies) and may be a specific isotype or only light or heavy
chain molecules.
Increased serum viscosity is a result of these diseases and can
interfere with testing. The increased concentrations of serum
proteins can cause nonspecific aggregation (as opposed to
agglutination) of erythrocytes called rouleaux, which is seen as a
stacking of RBCs, like a stacking of coins. It often occurs in multiple
myeloma patients.
If rouleaux is suspected, the saline replacement technique may be
needed to distinguish true cell agglutination from nonspecific
aggregation.

Autoimmune Disease
Autoantibodies are produced against the host’s own cells and
tissues.
It is unknown why this loss of tolerance to self-antigens occurs, but
there are many possible explanations such as aberrant antigen
presentation, failure to obtain clonal deletion, anti-idiotypic network
breakdown, and cross reactivity between self and non self -antigens.
Autoimmune hemolytic anemias are an important problem in
testing and transfusion
#MLSWATER2022 Page 10 of 10
MODULE 1: Introduction to Immunohematology (part 5)
MLS 18, February 23, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
(S) strain, which has a smooth polysaccharide capsule, kills mice through
Legend: pneumonia.
Notes/Module Packet Bullet o The nonvirulent rough (R) strain lacks the outer capsule and is
Reference Book nonlethal to mice.

MODULE OUTLINE 1944, Oswald T. Avery and his group


I. Historical Background and the current trends in Transfusion Medicine o at Rockefeller Institute were able to reproduce this transformation
II. Blood Preservation in vitro.
III. Review of Basic Genetics
IV. Review of Fundamentals of Immunology Max Delbrück of Vanderbilt University, Salvador Luria of Indiana
University, and Alfred Hershey of Carnegie’s Department of Genetics
V. Review of Molecular Biology
at Cold Spring Harbor
A. DNA: The Genetic Material
o Confirmation that DNA was the genetic material came from the
B. Nucleic Acid Testing
“phage group”.
C. Central Dogma
o Coincided in their interest to research bacteriophages, viruses that
1. Replication
infect bacteria.
2. Transcription
o Bacteriophages are very small viruses that consist only of a DNA
3. Translation
core enclosed in a protein capsule. It was known that, during
bacterial infection, the phage particles reproduce inside the cell and
LEARNING OUTCOMES
lyse the bacteria to release a new generation of viruses.
At the end of this module, the student shall be able to:
1. Explain how DNA was proven to be the carrier of genetic
Alfred Hershey and Martha Chase
information.
o In 1952, they reported an experiment that convinced the scientific
2. Explain the central dogma of molecular biology.
community that DNA was indeed the genetic material.
3. Describe the basic mechanism replication, transcription, and
translation.
DNA STRUCTURE
4. Explain how nucleic acid testing (NAT) in blood donor improves the
process of screening for infectious disease. By 1950, the chemical composition of nucleic acids was known.
o Nucleic acids were known to be long molecules composed of three
distinct chemical subunits:
DNA: THE GENETIC MAT ERIAL  Five-carbon sugar
1928, Fred Griffith  Acidic phosphate
o A British microbiologist that presented a model system that was key  Four types of nitrogen-rich bases.
to demonstrating that DNA is the genetic material. o Two forms of nucleic acids were differentiated by their sugar
o He used two naturally occurring strains of pneumococcus composition: RNA contained ribose and DNA 2-deoxyribose.
bacterium that differed in their infectivity of mice. The virulent smooth

#MLSWATER2022 Page 1 of 5
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o Both possessed adenine, guanine, and cytosine. RNA contained Expression of Genetic Information
uracil, and DNA contained thymine. Genes act by determining the sequence of amino acids and therefore
the structure of proteins.
1950, Erwin Chargaff of Columbia University Knowledge of the structure of DNA revealed that the genetic
o He reported a consistent one-to-one ratio of adenine to information must be specified by the order of the four bases (A, C, G,
thymine and guanine to cytosine in DNA samples from and T).
different organisms. Proteins are polymers of 20 amino acids, the sequence of which
determines their structure and function.
1951, Maurice Wilkins and Rosalind Franklin Because DNA was in the nucleus, separated from the cytoplasm by the
o He produced x-ray diffraction photographs of DNA, which nuclear membrane, an intermediary molecule had to be responsible
suggested a helical molecule with repeats of 34 angstroms for conveying the genetic information from the nucleus to the
(Å) and a width of 20 Å. cytoplasm.
RNA was a good candidate for this role. Its structure suggested that
1953, James Watson (trained in the phage group) and Francis RNA could be produced from a DNA template and that RNA was
Crick (a physicist trained in x-ray crystallography, published a located mainly in the cytoplasm, where protein synthesis occurred.
paper in the journal Nature in which they assembled the
puzzle pieces of DNA structure.)
o They proposed that the DNA molecule was a helix. DNA was THE CENTRAL DOGMA OF MOLECULAR BIOLOGY
a helical ladder, the rails of which were built from alternating The Central Dogma of Molecular Biology states that: DNA makes RNA
units of deoxyribose and phosphate. makes proteins.
o Each rung of the ladder was composed of a pair of Deciphering the genetic code confirmed the central dogma of
nucleotides (a base pair) held together by hydrogen bonds. molecular biology first formulated by Francis Crick.
The double helix consisted of two strands of nucleotides that The genetic material is DNA.
ran in opposite directions (antiparallel). DNA is self-replicating and is transcribed into mRNA, which in turn
o Consistent with the 34-Å repeat from x-ray diffraction, serves as a template for the synthesis of proteins.
10 base pairs were stacked on top of each other at Although Crick’s central dogma remains true, the knowledge acquired
each turn of a helix. in the following
o In agreement with Chargaff’s observation, adenine always years has refined and enlarged it, a process that continues now and
paired with thymine, and guanine always paired with into the future.
cytosine. Thus the nucleotide alphabet of one half of the
DNA helix determined the alphabet of the other half. Scientists have added further qualifications to the central dogma:
1. Genes are not “fixed.”
DNA FUNCTION 2. DNA sequences and protein amino acid sequences are not entirely
Genes must be replicated and passed down to each new cell and each collinear.
3. Some RNA molecules display catalytic activity; by “RNA
interference,” small RNA molecules help to regulate gene
expression.

new generation.
Genes must also provide the information for protein synthesis.
#MLSWATER2022 Page 2 of 5
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
DNA REPLICATION DNA TRANSLATION
The Watson-Crick structure revealed how DNA could replicate. The mRNA formed in transcription is transported out of the nucleus,
The DNA molecule is made of two antiparallel complementary strands. into the cytoplasm, to the ribosome (the cell's protein synthesis
Cytosine always pairs with guanine, and thymine always pairs with factory). Here, it directs protein synthesis.
adenine. Messenger RNA is not directly involved in protein synthesis − transfer
The information in one DNA strand predicts the information in the RNA (tRNA) is required for this.
other strand. During replication, the hydrogen bonds break, the The process by which mRNA directs protein synthesis with the
strands separate, and each one functions as template for the synthesis assistance of tRNA is called translation.
of another complementary half molecule.
Two identical DNA molecules are generated, each containing an Recombinant DNA
original strand and a new complementary strand, each to be passed to The big breakthrough came with the development of recombinant
a daughter cell DNA technology.
Because each daughter double helix contains an “old” strand and a o DNA from one organism, humans for example, can be
newly synthesized strand, this model of replication is called “cut and pasted” into a carrier DNA molecule or vector.
semiconservative.
o The new DNA molecule, which is a “recombinant” of the
original DNA with the vector DNA, can be introduced into
DNA TRANSCRIPTION another, usually simpler, host organism. Because the
Transcription is the process by which DNA is copied (transcribed) to genetic code is almost universal, the host organism treats
mRNA, which carries the information needed for protein synthesis. the gene as its own. This technique is called molecular
Transcription takes place in two broad steps. cloning.
o First, pre-messenger RNA is formed, with the involvement of o Cloning is the reproduction of daughter cells from one
RNA polymerase enzymes. The process relies on Watson-Crick single cell by fission or mitotic division, giving rise to a
base pairing, and the resultant single strand of RNA is the population of genetically identical clones.
reverse-complement of the original DNA sequence. o In DNA cloning, the DNA fragment of interest
o The pre-messenger RNA is then "edited" to produce the desired carried by the vector is introduced into a host
mRNA molecule in a process called RNA splicing. cell. Successive divisions of the host cell create a
population of clones containing the DNA
Reverse transcription fragment of interest.
o RNA is "reverse transcribed" into DNA
o catalyzed by reverse transcriptase enzymes, allows retroviruses, DETECTION OF NUCLEIC ACIDS AND PROTEINS
including the human immunodeficiency virus (HIV), to use RNA Nucleic Acid Hybridization
as their genetic material Base pairing between complementary strands of DNA or RNA allows
o Reverse transcriptase enzymes have also found applications in the specific detection of nucleic acid sequences.
biotechnology, allowing scientists to convert RNA to DNA for Double-stranded nucleic acids denature at high temperature (90⁰ to
techniques such as PCR. 100⁰C) and renature when cooled to form double-stranded molecules
as dictated by complementary base pairing.
Nucleic acid hybrids can be formed between two strands of DNA, two
strands of RNA, or one strand of RNA and one of DNA. DNA or RNA

#MLSWATER2022 Page 3 of 5
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
sequences complementary to any purified DNA fragment can be
detected by nucleic acid hybridization. o Fluorescent in situ Hybridization (FISH)
o Southern Blotting  In this technique, scientists use fluorescent probes to
 In this technique developed by E. M. Southern, the DNA to detect homologous DNA or RNA sequences in
be analyzed is digested with one or more restriction chromosomes or intact cells.
endonucleases and the fragments separated by agarose gel  In this case, the hybridization of the probe to specific cells
electrophoresis. or sub-cellular structures is determined by direct
 The gel is then placed over a nitrocellulose or nylon microscopic examination.
membrane and overlaid with transfer buffer. The DNA
fragments are transferred or “blotted” when vacuum is PCR-Based Techniques
applied by the flow of transfer buffer. Amplification of DNA by PCR allows the detection of even single
 The membrane-bound fragments have the same relative copies of DNA molecules.
positions as the fragments separated by size on the gel. The specificity of the PCR amplification depends on the primers that
hybridize to complementary sequences of the template molecule
o Northern Blotting spanning the target DNA fragment.
 This technique is a variation of the Southern blotting and is With carefully chosen primers, PCR can be used to selectively
used for detection of RNA instead of DNA. amplify DNA molecules from complex mixtures, such as genomic
 Total cellular RNAs are extracted and fractioned by size DNA from cell extracts.
through gel electrophoresis. o Reverse Transcription-PCR (RT-PCR)
 The RNAs are then blotted onto a filter and detected by  By adding a step of cDNA synthesis by reverse
hybridization with a labeled probe. This technique is used transcription (RT) prior to the PCR amplification, single
for studying gene expression. copies of RNA can be detected. By RT-PCR,
 RNA molecules can be specifically amplified from total
o DNA microarrays RNA obtained from cell extracts or tissue sections.
 also called gene chips, allow tens of thousands of genes to
be analyzed simultaneously. o Real-Time PCR in real-time PCR (also called RT-PCR)
 A gene chip consists of a glass slide or a membrane filter  the product formed in each cycle of amplification is
onto which oligonucleotides or fragments of cDNA are detected by fluorescence at the same time that it is
printed by a robot in small spots at high density. produced.
 Because a chip can contain more than 10,000 unique DNA  Combined with reverse transcription, this method allows
sequences, scientists can produce DNA microarrays the quantification of specific mRNA in complex mixtures
containing sequences representing all the genes in a and is extremely useful in the study of gene expression.
cellular genome.  the high sensitivity of PCR-based DNA and RNA detection
 A common application of this technique is the study of techniques makes them valuable for early detection of
differential gene expression; for example, the comparison transfusion-transmitted viruses, such as HIV and hepatitis
of the genes expressed by a normal cell as opposed to B and C.
those of a tumor cell.  Nucleic acid testing (NAT) is rapidly becoming a standard
method in blood banks. It allows the detection of

#MLSWATER2022 Page 4 of 5
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
pathogens before the appearance of a testable immune
response, such as screening of antibodies.
 Reducing the “window” period (during which donors can
be infected but do not yet test positive) helps to enhance
the safety of blood products.

Antibodies as Probes for Proteins


Gene expression at the protein level can be studied by using labeled
antibodies as probes. In particular, monoclonal antibodies are
widely used in the technique called immunoblotting.

Recombinant DNA Libraries


A way of isolating single genes is by the production of recombinant
DNA libraries. Instead of trying to fish one gene out of a mass of
genomic DNA or cDNA, each gene is physically separated and
introduced into a vector.
The target gene is selected by screening each of the individual
pieces with a specific probe. Recombinant DNA libraries are
collections of clones that contain all genomic or mRNA sequences of
a particular cell type.
Clones containing a specific gene are identified by hybridization with
a labeled probe, such as a cDNA or genomic clone or a PCR product.

Red Cell Genotyping


In cases where serology is not possible or not sensitive enough or
where discrepancies occur, genotyping provides results that can be
used to obtain a blood type on a donor or patient.

Applications for genotyping:


1. Fetal DNA typing
2. Blood group typing of donors for alloimmunized patients
3. Screening blood donors to locate rare blood group phenotypes
4. Screening blood inventory for antigen negative units
5. Determining the frequency of blood group polymorphism
6. Determining zygosity for the fathers of fetuses at risk for HDFN
7. Blood group typing of patients with autoimmune hemolytic anemia
and other disease.

-END OF TRANSCRIPTION-
#MLSWATER2022 Page 5 of 5
MODULE 2&3: Rh Naming
MLS 18, March 13, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`

SUMMARY OF BLOOD GROUP SYSTEMS Note: “p” refers to short arm of chromosome; “q” refers to long arm of
ISBT # BLOOD GROUP CHROMOSOME NUMBER
For Rh Blood group
001 ABO 9q Chr1: RHD gene: D polypeptide; RHCE gene: RHCe,RHcE,RHce,RHCE
002 MNS 4q polypeptides Chr 6: RHAG: Rh associated glycoprotein that forms complexes
003 P 22q with Rh polypeptides
004 Rh 1p
005 Lutheran 19q Rh Nomenclatures
006 Kell 7q 1. Fisher-Race: genetics and serology
007 Lewis 19p 2. Wiener: shorthand
008 Duffy 1q 3. Rosenfield: presence or absence of a given antigen
009 Kidd 18q 4. ISBT: catalogues each antigen within a blood group
system
010 Diego 17q
011 Cartwright 7q Fisher-Race Terminology
012 Xga Xp o Based on closely linked alleles D, C/c, and E/e
013 Scianna 1p o d is an amorph and does not produce a phenotypic product
014 Dombrock 12p o d= absence of D antigen
015 Colton 7p
016 Landsteiner-Weiner 19p Wiener terminology
017 Chido-Rogers 6p • Wiener is the “shorthand” version of Fisher-Race
018 H 19q o R= presence of D
019 Kx Xp o r= d, or absence of D antigen
o 1 or single prime= presence of C
020 Gerbich 2q
o 2 or doubleprime= presence of E
021 Cromer 1q
022 Knops 1q Antigen Wiener
023 Indian 11p D R
024 Ok 19p D r
025 Raph 11p C 1 or ‘
026 John-Milton Hagen 15q E 2 or “
027 I 6p
028 Globoside 3q
029 GIL 9p
#MLSWATER2022 Page 1 of 2
MODULE 2-3- RH NAMING
Fisher-Race Wiener Sample Question:
Rh positive Dce R0
Question 1
DCe R1 • A patient’s red blood cells are tested for the following Rh antigens:
DcE R2 Anti-D : +
DCE Rz Anti-C : +
Rh negative dce r Anti-E: 0
dCe r’ anti-c: +
dcE r” anti-e: +
dCE y
Antigens present: D, C, c, e
Rosenfield Most likely genotype: DCe/dce R1r
o This system simply describes the presence or absence of the antigen Other possibilities: Dce/dCe R0r’
on the RBC. There is no genetic basis. DCe/Dce R1R0
o D=1, C=2, E=3, c=4, e=5 Question 2
o Example: R1r (DCe/dce): Rh:1,2,-3,4,5 • A patient’s red blood cells are tested for the following Rh antigens:
• E is number 3; E antigen is not present and is therefore designated with Anti-D: 0
-3 Anti-C: 0
ISBT Anti-E: 0
o International Society of Blood Transfusion Numeric Terminology. anti-c: +
o Rh blood group is assigned the prefix 004 anti-e: +
o Each antigen assigned to the Rh blood group is given a unique
number to complete the six-digit number. Antigens present: c, e
o Example: E antigen 004003 Most likely genotype: dce/dce rr
o Advantage over Rosenfield is that it is a purely numeric system, Other possibilities: None
which is easier for data processing.
Question 3
• A patient’s red blood cells are tested for the following Rh antigens:
Anti-D: +
Anti-C: +
Anti-E: 0
anti-c: 0
anti-e: +

Antigens present: D, C, e
Most likely genotype: DCe/DCe R1R1
Other possibilities: DCe/dCe R1r’

#MLSWATER2022 Page 2 of 2
MODULE 2 Part 1: The Anti-globulin Test
MLS 18, March 5, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
ANTI-GLOBULIN TEST
Legend: Antiglobulin test (also called Coombs’ test)
Transcription Bullet Based on the principle that: anti-human globulins (AHGs) obtained
Notes/Module Packet Bullet from immunized nonhuman species bind to human globulins such as
IgG or complement, either free in serum or attached to antigens on red
MODULE OUTLINE blood cells (RBCs).
I. Anti- Globulin Test There are two major types of blood group antibodies, IgM and IgG.
A. History o Because of their large pentamer structure, IgM antibodies bind to
B. AHG Reagents corresponding antigen and directly agglutinate RBCs suspended in
1. Polyspecific AHG saline.
2. Monospecific AHG o IgG antibodies are termed non-agglutinating because their
C. Preparation of AHG monomer structure is too small to agglutinate sensitized RBCs
D. Antibodies required in AHG directly.
E. Principle of the Anti-Globulin Test The addition of AHG containing anti-IgG to RBCs sensitized with IgG
F. Direct and Indirect Anti-Globulin Test antibodies allows for hemagglutination of these sensitized cells. Some
G. Factors Affecting the Anti-Globulin Test blood group antibodies have the ability to bind complement to the RBC
H. Modified and Automated Antiglobulin Test Techniques membrane. Antiglobulin tests detect IgG and/or complement-sensitized
I. Use of Polyspecific Versus Monospecific AHG in the IAT RBCs.

LEARNING OUTCOMES HISTORY


1. State the principle of antiglobulin test. Before the discovery of the antiglobulin test, only IgM antibodies had
2. Differentiate monoclonal from polyclonal and monospecific from been detected.
polyspecific antihuman globulin (AHG) reagents. The introduction of the antiglobulin test permitted the detection of non-
3. Compare and contrast the indirect antiglobulin test (IAT) and the agglutinating IgG antibodies and led to the discovery and
direct antiglobulin test (DAT). characterization of many new blood group systems.
4. Identify the factors that affect the antiglobulin test.
5. Formulate the steps necessary to resolve errors associated with In 1945, Coombs and associates
performance of the antiglobulin test. o Described the use of the antiglobulin test for the detection of weak
and nonagglutinating Rh antibodies in serum.

In 1946, Coombs and coworkers


o Described the use of AHG to detect in-vivo sensitization of the RBCs
of babies suffering from hemolytic disease of the newborn (HDN).

#MLSWATER2022 Page 1 of 6
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o The first of the Kell blood group system antibodies and the
associated antigen were reported only weeks after Coombs had MONOSPECIFIC AHG
described the test. Contain only one antibody specificity: either anti-IgG or antibodies to
specific complement components such as C3b or C3d.
In 1908, Moreschi Licensed monospecific AHG reagents in common use are anti-IgG and
o Described the Principle of Coombs test. anti-C3bC3d.
o Moreschi’s studies involved the use of rabbit antigoat serum to
Anti-IgG Reagent
agglutinate rabbit RBCs that were sensitized with low non-
Reagents labeled anti-IgG contain no anticomplement activity.
agglutinating doses of goat anti-rabbit RBC serum. The antiglobulin
Anti-IgG reagents contain antibodies specific for the Fc fragment of
test can be used to detect RBCs sensitized with IgG alloantibodies,
the gamma heavy chain of the IgG molecule.
IgG autoantibodies, and complement components.
If not labeled “gamma heavy chain–specific,” anti-IgG may contain
anti–light chain specificity and therefore react with cells sensitized
How does the AHG reagent was produced?
with IgM and IgA as well as with IgG.
o It involves the injection of Human Serum into the Rabbit to
produce an Anti-human Serum.
Anti-Complement
o After the adsorption, to remove the heterospecific antibodies and
Anti-complement reagents, such as anti-C3b-C3d reagents, are
the dilution to avoid the Prozone Effect
reactive against the designated complement components only and
 Prozone Effect- excessive Antibodies
contain no activity against human immunoglobulins.
o AHG Serum still remains sufficient activity to permit cross linking of
the adjacent RBCs synthesize with IgG Antibodies.
 This can later on produce the heme agglutination. PREPARATION OF AHG
POLYCLONAL ANTIBODIES are a mixture of antibodies from different
 AHG able to detect in vivo and in vitro sensitization of RBCs plasma cell clones. The resulting polyclonal antibodies recognize
o In vivo: Direct Antiglobulin Test (DAT) different antigenic determinants (epitopes), or the same portion of the
 One-stage procedure antigen but with different affinities. Hybridoma technology can be used
o In vitro: Indirect Antiglobulin Test (IAT) to produce monoclonal antiglobulin serum.
 Two-stage procedure
MONOCLONAL ANTIBODIES are derived from one clone of plasma cells
AHG REAGENTS and recognize a single epitope.
POLYSPECIFIC AHG
Contains antibody to human IgG and to the C3d component of human
PREPARATION OF POLYSPECIFIC AHG
complement. POLYCLONAL AHG PRODUCTION
Other anticomplement antibodies, such as anti-C3b, anti-C4b, and anti- Polyclonal AHG is usually prepared in rabbits, although when large
C4d, may also be present. volumes of antibody are required, sheep or goats may be used. In
Commercially prepared polyspecific AHG contains little, if any, activity contrast with the early production methods, in which a crude globulin
against IgA and IgM heavy chains. fraction of serum was used as the immunogen, modern production
However, the polyspecific mixture may contain antibody activity to commences with the purification of the immunogen from a large pool of
kappa and lambda light chains common to all immunoglobulin classes, normal sera.
thus reacting with IgA or IgM molecules.

#MLSWATER2022 Page 2 of 6
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
MONOCLONAL AHG PRODUCTION Anti-Complement
The monoclonal antibody technique devised by Kohler and Milstein has Some antibodies “fix” complement components to the RBC membrane
been used to produce AHG and has proved particularly useful in after complexing of the antibody with its corresponding antigen.
producing high-titer antibodies with well-defined specificities to IgG and These membrane-bound complement components can be detected by
to the fragments of C3. the anticomplement activity in AHG.
Monoclonal antibody production begins with the immunization of
laboratory animals, usually mice, with purified human globulin. Use of Polyspecific Versus Monospecific AHG in the IAT
After a suitable immune response, mouse spleen cells containing
Petz and coworkers
antibody-secreting lymphocytes are fused with myeloma cells.
Compared monospecific anti-IgG with polyspecific AHG.
The resulting “hybridomas” are screened for antibodies with the
required specificity and affinity. The antibody-secreting clones may then They also compared the albumin technique with low ionic strength
be propagated in tissue culture or by inoculation into mice, in which solutions (LISS)-suspended RBCs.
case the antibody is collected as ascites. Four Jka antibodies were detected with polyspecific but not with
monospecific anti-IgG using albumin or LISS-suspended RBCs.
PREPARATION OF MONOSPECIFIC AHG An additional anti-Jka was detected only with polyspecific AHG when
using LISS but not with albumin.
It is prepared by a production process similar to that described for Also, five antibodies of antiKell, anti-Jka, and Fya specificities were
polyspecific AHG; however, it contains only one antibody specificity. detected when using LISS, but not albumin, with both polyspecific AHG
Monospecific anti-IgG is usually of polyclonal origin; however, and antiIgG.
monoclonal anti-IgG has been prepared effectively by hybridoma o Their results concluded that some clinically significant antibodies
technology. are detected with the anticomplement component of AHG but not
Monospecific anticomplement reagents are often a blend of monoclonal with anti-IgG.
anti-C3b and monoclonal anti-C3d. Also determined the number of false-positive reactions obtained when
using polyspecific AHG versus anti-IgG with LISS and albumin.
ANTIBODIES REQUIRED IN AHG o False-positive reactions were defined as those caused by antibodies
Anti-IgG with no definable specificity or by antibodies considered to be
AHG must contain antibody activity to non-agglutinating blood group clinically insignificant because of optimum reactivity at cold
antibodies. The majority of these antibodies are a mixture of IgG1 and temperatures (anti-I, anti-H, anti-P1, anti-M).
IgG3 subclass. o Of the unwanted positive reactions, 93% were shown to be caused
Rarely, non-agglutinating IgM antibodies may be found; however, they by C3 on the cells.
have always been shown to fix complement and may be detected by
anticomplement. Engelfriet and others
IgA antibodies with Rh specificity have been reported; however, IgG have also shown that degradation of C3b to C3d can occur in vitro,
antibody activity has always been present as well. providing that the incubation period is greater than 1 hour.
The only RBC alloantibodies that have been reported as being solely IgA In 1976, Garratty and Petz confirmed the need for anti-C3d activity in
have been examples of anti-Pr,12 and those antibodies were AHG for use in the DAT. They also confirmed Engelfriet’s observation
agglutinating. that, given sufficient time, cell-bound C3b could be degraded to C3d in
IgA autoantibodies have been reported, although very rarely. vitro.
o Therefore, anti-IgG activity must be present in the AHG reagent.

#MLSWATER2022 Page 3 of 6
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
NOTE: Positive results are monitored by a DAT panel using monospecific anti-
The detection of C3d on the RBC membrane is important in the IgG and anti-C3d to determine the specific type of protein sensitizing
investigation of both warm and cold autoimmune hemolytic anemia the cell.
(AIHA). Many cases of warm AIHA are associated with both IgG and The saline control serves to detect spontaneous agglutination of cells
C3d coating the RBCs. In cold AIHA, C3d may be the only globulin or reactions occurring without the addition of AHG reagents.
detectable on the RBC. In warm AIHA, including drug-induced hemolytic anemia, the RBCs
In the investigation of AIHA, a DAT is performed initially with may be coated with IgG or C3d, or both.
polyspecific AHG. If globulins are detected on the RBC membrane,
follow-up testing with monospecific AHG (antiIgG, anti-C3d) is
performed to identify the coating proteins. Although the RBCs of
most patients with AIHA are coated with IgG, the cells of some
patients will exhibit both IgG and complement coating or
complement alone. The presence of complement alone may support
the diagnosis of AIHA, rendering the finding significant.

PRINCIPLES OF THE ANTIGLOBULIN TEST


1. Antibody molecules and complement components are globulins.
2. Injecting an animal with human globulin stimulates the animal to
produce antibody to the foreign protein (i.e., AHG). Serologic tests
employ a variety of AHG reagents reactive with various human
globulins, including anti-IgG, antibody to the C3d component of
For the case of HDN:
human complement, and polyspecific reagents that contain both anti-
o The RBC of the Fetus contains antigens to its surface. This will bind
IgG and anti-C3d activity.
to the Antibodies formed by the mother
3. AHG reacts with human globulin molecules, either bound to RBCs or
o After the Antigen-Antibody binding, the Anti-IgG found in Coomb’s
free in serum.
Reagent will bind itself to the Antibody of the mother.
4. Washed RBCs coated with human globulin are agglutinated by AHG.
o This will lead to the formation of agglutination.

DAT (Direct Antiglobulin Test)


Evaluation of Positive DAT
Detects in-vivo sensitization of RBCs with IgG and/or complement
 The American Association of Blood Banks Technical Manual states that:
components.
“The results of serological tests are not diagnostic; their significance can
Clinical conditions that can result in in-vivo coating of RBCs with
only be assessed in relationship to the patient’s clinical condition.
antibody and/or complement are:
o Hemolytic disease of the newborn (HDN)
o Hemolytic transfusion reaction (HTR) IAT (Indirect Antiglobulin Test)
o Autoimmune and drug-induced AIHA. The IAT is performed to determine in-vitro sensitization of RBCs
Initial DATs include testing one drop of a 3 to 5 percent suspension of Used in the following situations:
washed RBCs with polyspecific (anti-IgG, anti-C3d) reagent. o Detection of incomplete (non-agglutinating) antibodies to
potential donor RBCs (compatibility testing) or to screening
cells (antibody screen) in serum
#MLSWATER2022 Page 4 of 6
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
o Determination of RBC phenotype using known antisera (e.g., MODIFIED AND AUTOMATED ANTIGLOBULIN TEST TECHNIQUES
Kell typing, weak D testing)
Low Ionic Polybrene Technique
o Titration of incomplete antibodies
In 1980, Lalezari and Jiang reported on the adaptation of the automated
low ionic polybrene (LIP) technique for use as a manual procedure.
The technique relies on low ionic conditions to rapidly sensitize cells
with antibody.
Polybrene, a potent rouleaux-forming reagent, is added to allow the
sensitized cells to approach each other to permit cross-linking by the
attached antibody.
A high ionic strength solution is then added to reverse the rouleaux;
however, if agglutination is present, it will remain. The test can be
carried through to an AHG technique if required.
If this is performed, a monospecific anti-IgG reagent must be used
because the low ionic conditions cause considerable amounts of C4 and
C3 to coat the cells and would give false-positive reactions if a
polyspecific reagent were used.

Enzyme-Linked Antiglobulin Test (ELAT)


In IAT: There are Two Stages involved: An RBC suspension is added to a microtiter well and washed with saline.
o Stage 1 AHG, which has been labeled with an enzyme, is added.
 We have the serum of the patient, it contains free Antibody, The enzyme labeled AHG will bind to IgG-sensitized RBCs. Excess
this will be collected and allowed to be mixed with the blood antibody is removed, and enzyme substrate is added.
of the donor. The amount of color produced is measured spectrophotometrically and
 The Antigen present at the surface of the donor’s blood will is proportional to the amount of antibody present.
bind to the Antibody of the patient. The optical density is usually measured at 405 nm. The number of IgG
o Stage 2 molecules per RBC can also be determined from this procedure.
 After the Antibody-Antigen binding, the Coomb’s Reagent
containing Anti-IgG will now bind to the Antibody present in Solid Phase
the patient.
Solid-phase technology may be used for the performance of antiglobulin
FACTORS AFFECTING THE ANTIGLOBULIN TEST tests. Several different techniques have been reported using either test
1. Ratio of serum to cells. tubes or microplates.
2. Reaction medium. With the availability of microplate readers, this modification lends itself
3. Temperature. to the introduction of semi-automation.
4. Incubation time Direct and indirect tests can be performed using solid-phase
5. Washing of RBC methodology.
6. Addition of AHG o In the former, antibody is attached to a microplate well, and RBCs
7. Centrifugation for reading are added. If antibody is specific for antigen on RBCs, the bottom of
-Refer to reference book for in depth explanation

#MLSWATER2022 Page 5 of 6
MODULE 1- INTRODUCTION TOIMMUNOHEMATOLOGY
the well will be covered with suspension; if no such specificity
occurs, RBCs will settle to the bottom of the well.
o In the latter, known RBCs are bound to a well that has been treated
with glutaraldehyde or poly L-lysine. Test serum is added to RBC-
coated wells, and if antibody in serum is specific for antigen on fixed
RBCs, a positive reaction occurs.

Anti-Complement
The gel test is a process to detect RBC antigen-antibody reactions by
means of using a chamber filled with polyacrylamide gel. The gel acts as
a trap; free unagglutinated RBCs form pellets in the bottom of the tube,
whereas agglutinated RBCs are trapped in the tube for hours.
Therefore, negative reactions appear as pellets in the bottom of the
microtube, and positive reactions are fixed in the gel.
There are three different types of gel tests: neutral, specific, and
antiglobulin.
o A neutral gel does not contain any specific reagent and acts only by
its property of trapping agglutinates. The main applications of
neutral gel tests are antibody screening and identification with
enzyme-treated or untreated RBCs and reverse ABO typing.
o Specific gel tests use a specific reagent incorporated into the gel
and are useful for antigen determination.

-END OF TRANSCRIPTION-

#MLSWATER2022 Page 6 of 6
MODULE 2 Part 2: ABO BLOOD GROUP SYSTEM
MLS 18, March 5, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo` 11. Interpret the results from an ABO typing and resolve any
discrepancies, if present.
Legend:
Transcription Bullet
Notes/Module Packet Bullet BLOOD GROUP
MODULE OUTLINE Classification of blood based on inherited differences (polymorphisms)
I. Blood Group in antigens on the surfaces of the red blood cells
II. ABO Antigens Inherited differences of white blood cells (leukocytes), platelets
III. ABO Antibodies (thrombocytes), and plasma proteins also constitute blood groups.
IV. Inheritance of the ABO Blood Groups The human ABO blood groups were discovered by Austrian-born
V. ABO Subgroups American biologist Karl Landsteiner in 1901.
VI. Bombay Phenotype o Landsteiner found that there are substances in the blood, antigens
VII. Effects of Disease on the Expression of ABH Antigens and Antibodies and antibodies that induce clumping of red cells when red cells of
VIII. Laboratory tests to detect and identify ABO Antigens and Antibodies one type are added to those of a second type.
IX. ABO Discrepancies o He recognized three groups—A, B, and O—based on their reactions
to each other. He was inadvertently the first individual to perform
LEARNING OUTCOMES forward and reverse grouping.
1. Describe the reciprocal relationships between ABO antigens and
antibodies for blood groups O, A, B, and AB. Forward grouping (front type)
2. Explain the effects of age on the production of isoagglutinins. Defined as using known sources of commercial antisera (anti-A, anti-
3. Describe the immunoglobulin classes of ABO antibodies in group O, A, B) to detect antigens on an individual’s RBCs.
and B individuals. Reverse grouping (back type)
4. Predict the ABO phenotypes and genotypes of offsprings from various Defined as detecting ABO antibodies in the patient’s serum by using
ABO mating. known reagent RBCs, namely A1 and B cells.
5. Explain the formation of H, A, and B antigens on the red blood cells
from precursor substance to immunodominant sugars. AB Group
6. Describe the qualitative and quantitative differences between A1 and Was identified a year later by another research team
A2 phenotypes. Red cells of the A group clump with donor blood of the B group; those
7. Describe the reactivity of Ulexeuropaeus with the various ABO of the B group clump with blood of the A group; those of the AB
groups. group clump with those of the A or the B group because AB cells
8. Describe the characteristics of the weak subgroups of A (A3, Ax, Aend, contain both A and B antigens
Am, Ay, Ael) and of the Bombay phenotypes. Those of the O group do not generally clump with any group, because
9. Explain the effects of disease on the expression of ABH antigens and they do not contain either A or B antigens.
antibodies.
10. Perform laboratory tests to detect and identify ABO antigens and
antibodies.
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
ABO forward and reverse grouping tests must be performed on all o The combination of pregnancy and transfusion is a particularly
donors and patients. ABO grouping is the most frequently performed potent stimulus.
test in the blood bank. Individual blood group antigens vary in their antigenic potential
There is always an inverse reciprocal relationship between the o For example, some of the antigens belonging to the Rh and ABO
forward and reverse type; thus, one serves as a check on the other. systems are strongly immunogenic (i.e., capable of inducing
antibody formation), whereas the antigens of the Kidd and Duffy
IMPORTANCE OF ANTIGENS AND ANTIBODIES blood group systems are much weaker immunogens.
o The blood group antigens are not restricted solely to red cells or
The red cells of an individual contain antigens on their surfaces that
even to hematopoietic tissues.
correspond to their blood group and antibodies in the serum that
identify and combine with the antigen sites on the surfaces of red cells The antigens of the ABO system are widely distributed throughout the
of another type. tissues and have been unequivocally identified on platelets and white
cells (both lymphocytes and polymorphonuclear leukocytes) and in skin,
Agglutination the epithelial (lining) cells of the gastrointestinal tract, the kidney, the
urinary tract, and the lining of the blood vessels.
The reaction between red cells and corresponding antibodies that
Evidence for the presence of the antigens of other blood group systems
cause the clumping of RBC
Agglutinogens on cells other than red cells is less well substantiated.
Antigens on the surfaces of red cells responsible for agglutination Among the red cell antigens, only those of the ABO system are
regarded as tissue antigens
o Therefore, need to be considered in organ transplantation.
Antibodies are part of the circulating plasma proteins known as
immunoglobulins, which are classified by molecular size and weight and
ABO ANTIGENS
by several other biochemical properties.
Located on the surface of the red blood cell.
Most blood group antibodies are found either on immunoglobulin G
Also present on lymphocytes, thrombocytes, organs, endothelial cells,
(IgG) or immunoglobulin M (IgM) molecules,
and epithelial cells.
o Occasionally the immunoglobulin A (IgA) class may exhibit blood
group specificity. First detected by Landsteiner as he performed his mixing tests
Naturally occurring antibodies are the result of immunization by The biochemistry and structure of ABO antigens are well-established.
substances in nature that have structures similar to human blood Antigens of the ABO system are well-developed in adults.
groups. o They are detectable at 5 to 6 weeks of gestation.
o Present in an individual despite the fact that there has been no Newborns demonstrate weaker antigens, but ABO antigens are fully
previous exposure to the corresponding red cell antigens developed by two to four years of age.
o Example: anti-A in the plasma of people of blood group B and anti-B One factor contributing to the difference in ABO antigen strength
in the plasma of people of blood group A. between newborns and adults is the number of branched
Immune antibodies are evoked by exposure to the corresponding red oligosaccharides.
cell antigen. o Adults demonstrate greater numbers of branched chains compared
Immunization (i.e., the production of antibodies in response to antigen) to newborns, who have more linear chains.
against blood group antigens in humans can occur as a result of o Adults have more branched chains and, hence, the ability to add on
pregnancy, blood transfusion, or deliberate immunization. more terminal sugars and produce more antigens.
o Newborns and infants have fewer antigen sites on their red cells.

#MLSWATER2022 Page 2 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
The branched chains permit attachment of more molecules to DEVELOPMENT OF H ANTIGEN
determine H antigen specificity. The H allele codes for the transferase, L-fucosyltransferase.
Following H antigen development, the A and/or B specific molecule may o This enzyme catalyzes the formation of the H antigen by transfer of
be attached. L-fucose to either type one or type two oligosaccharide chains.
o The L-fucose is the immunodominant sugar for the H antigen. It is
INHERITANCE OF ABH ANTIGENS the sugar that confers antigenic specificity to the H antigen.
As Bernstein discovered, ABO antigens are inherited in a simple The H antigen serves as a precursor for A and B antigens.
Mendelian fashion from an individual’s parents. The h allele is an amorph and does not produce a detectable product.
o Each individual possesses a pair of genes.
o Each gene occupies an identical locus on chromosome 9. DEVELOPMENT OF A AND B ANTIGENS
o There are three possible genes that can be inherited: A, B, and O. The H antigen oligosaccharide chain serves as a precursor for both the
A and B genes produce a detectable product while the O gene is an A and B antigens.
amorph that does not produce a detectable product. The A and B alleles each code for a transferase that attaches a sugar
o The expression of the A and B genes is codominant. molecule to the terminal end of the H antigen oligosaccharide chain,
The H antigen is required to produce A and/or B antigens. which forms either the A or B antigen.
o The H gene is also inherited in Mendelian fashion and occupies a o The A allele codes for N-acetylgalactosamine transferase.
locus on chromosome 19. o This transferase attaches N-acetyl-D-galactosamine to the H antigen
o Each parent contributes one gene, either H or h. forming the A antigen.
o The possible genetic combinations are HH, Hh, or hh. o The B allele codes for D-galactosyltransferase.
Individuals who are genetically either HH or Hh will produce the H o This transferase attaches D-galactose to the H antigen forming the B
antigen, and it can be detected on their red cells. antigen.
The frequency of occurrence of the H antigen in the Caucasian The product of the O allele is an enzymatically inactive protein.
population is greater than 99.99%. o Hence, this allele produces no detectable antigen (amorph)
Individuals inheriting an hh genotype do not produce the H antigen and Conversely, group O cells contain the most H antigen. This results from
have the Bombay Phenotype, Oh. no conversion of H antigen to A and/or B antigens.
Anti-H – Frequently demonstrated on the plasma of an individual with a o In comparison, group A1 B cells have the least amount of H antigen
Bombay phenotype since quantitatively the most H is converted to A1 and B antigens.

BIOCHEMICAL AND STRUCTURAL DEVELOPMENT OF ABH ANTIGENS ABO ANTIBODIES


Expression of A, B, and H genes does not result in the direct production “Antibodies directed against ABO antigens are the most important
of antigens. antibodies in transfusion medicine.”
o Rather, each gene codes for the production of an enzyme known as The ABO blood group presents a unique situation in
a transferase. Immunohematology.
Each transferase catalyzes the transfer of a carbohydrate molecule to an It is the only example of a blood group where each individual
oligosaccharide chain. The attached carbohydrate provides antigenic produces antibodies to antigens not present on the red cells.
specificity. These ABO antibodies were originally thought to be natural antibodies
formed with no apparent antigenic stimulus.
The O gene codes for an enzymatically inactive protein and, hence, no
antigen is produced.
#MLSWATER2022 Page 3 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
Since the antibodies are not stimulated by exposure to red cells, they o Anti-A,B is not required in forward grouping.
may also be considered non-red cell stimulated antibodies. Since it is a valuable reagent for determining subgroups of A and B, anti-
o However, some form of an antigenic stimulus must exist. A,B is often included as a routine part of forward grouping.
o The proposed mechanism is environmental. Monoclonal anti-A,B have replaced the use of human anti-A, B in
These “naturally occurring” substances resemble A and B antigens forward grouping.
and stimulate the production of complementary antibodies to the
antigens that are not present on the red cell surface. Anti-A1
As per Landsteiner’s Law, group B and O individuals produce anti-A.
NOTE: This anti-A can be separated by absorption procedures.
Newborns have no ABO antibodies. When newborns are tested, only These absorption procedures can produce two components of the
a forward group is performed. Newborns may exhibit passive ABO antibody found in group B and O individuals.
antibodies that have crossed the placental barrier. Reverse grouping o These components are anti-A and anti-A1.
of a newborn or umbilical cord serum indicates the blood group of o The anti-A1 antibody reacts specifically with A1 cells and not with
the mother. The child will begin antibody production, and have a A2 cells or cells from other subgroups of A.
detectable titer, at three to six months of age. ABO antibody Like other ABO antibodies, this antibody reacts optimally at room
production peaks at age five to ten years of age and continues in temperature or colder.
immunocompetent individuals throughout life. Titers begin to wane Anti-A1 is not considered clinically significant as it relates to transfusion.
in the elderly. o It is, however, significant when it causes incompatible crossmatches
at the immediate spin phase.
THE IMMUNOGLOBULIN CLASSES OF ABO ANTIBODIES IN GROUP ABO Antibodies to other A subgroups, such as A2, are not produced. These
IMMUNOGLOBULIN CLASS subgroups have the A antigen but in reduced amounts.
ABO antibodies are typically isoagglutinins. o Therefore, transfusion of A1 individuals with A2 cells will not
o They are saline agglutinins with optimal reactivity at 4°C. stimulate the production of anti-A2 since both A1 and A2 individuals
These naturally occurring antibodies are mostly IgM isotype have the A antigen in common.
o IgG and IgA classes of ABO antibodies have been detected.
The development of IgG antibodies occurs without apparent antigen CLINICAL SIGNIFICANCE OF ABO ANTIBODIES
exposure via transfusion of incompatible red cells or fetal maternal ABO antibodies are capable of causing both Hemolytic Disease of the
incompatibility. Fetus and Newborn (HDFN) and Hemolytic Transfusion Reactions
(HTR).
Anti-A,B These issues explain the clinical significance of “naturally occurring”
antibodies.
Group O individuals do not have A or B antigens on their cells.
HDFN usually presents itself with a maternal antibody of an IgG
o Consequently, they produce anti-A, anti-B, and anti-A,B.
isotype that corresponds to an antigen on the surface of the baby’s
Anti-A,B is an antibody that has cross- reactivity with A and B cells.
red cells.
This cross-reactive antibody detects a common molecular structure in The most common scenario is a group O mother and a group A baby.
both antigens. o ABO hemolytic disease may affect a woman’s first pregnancy.
o Although the antibody reacts with both antigens, it cannot be This is in contrast to Rh HDFN where the antigenic stimulation
divided into individual components (i.e. anti-A plus anti-B). usually occurs in the first pregnancy and subsequent antigen-
o Anti-A,B is used as a third antisera in forward grouping. positive newborns are affected.
#MLSWATER2022 Page 4 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
Hemolytic transfusion reaction occurs when a recipient is transfused ANTI-A AND ANTI-B
with red cells that are an ABO group incompatible with the antibodies Anti-A and anti-B are found in the sera of individuals who lack the
in his or her serum. Because of the complement-binding ability of the corresponding antigens.
ABO antibodies, this is always a life-threatening situation. They are produced in response to environmental stimulants, such as
As the recipient antibodies react with the incompatible red cells, bacteria, and have therefore been termed natural antibodies.
complement is activated and in vivo hemolysis, agglutination, and Antibody production begins after birth, reaching a peak at age 5 to
red blood cell destruction occurs. 10 and declining with increasing age.
ABO compatibility is also significant in solid organ transplantation. The antibodies formed to carbohydrate antigens are mostly
o For most organs, an ideal scenario for transplant is an ABO immunoglobulin M (IgM).
compatible solid organ. o IgM antibodies activate complement, which, in conjunction
o Post-transfusion antibody titer, and pheresis to reduce the with the high density of ABO antigen sites on RBCs,
titer of the incompatible antibody, will assist in achieving a o is responsible for the severe, life-threatening transfusion
positive outcome when an ABO incompatible organ is reactions that may be caused by ABO-incompatible
transplanted. transfusions.

EFFECTS OF AGE ON THE PRODUCTION OF ISOAGGLUTININS Hemolytic disease of the Newborn


Hemolytic disease of the newborn caused by ABO antibodies is
ISOAGGLUTININS usually mild, for the following reasons:
Isoagglutinins are antibodies produced by an individual that cause o placental transfer is limited to the fraction of IgG anti-A and
agglutination of RBCs in other individuals. anti-B found in maternal serum
People possess isoagglutinins directed toward the A or B antigen o fetal ABO antigens are not fully developed
absent from their own RBCs. o ABO tissue antigens provide additional targets for the
For example, type B or O individuals will usually possess anti-A. The antibodies.
anti-A is formed in response to exposure to A-like antigenic structures ABO-HDN is most often seen in non-group O infants of group O
found in ubiquitous non-red cell biologic entities (eg, bacteria). mothers, because anti-A, anti-B, and anti-A,B of group O mothers
often has a significant IgG component.
Isoagglutinins present in the newborn are passively acquired from
maternal circulation. Bombay Phenotype
o Such passively acquired isoagglutinins will gradually Potent anti-H (along with anti-A and anti-B) found in Oh (Bombay)
disappear, and the infant will begin to produce or para-Bombay nonsecretors destroys transfused RBCs of any ABO
isoagglutinins at 3 to 6 months of age. group, so these individuals must be transfused only with blood of
Isoagglutinins production may vary in patients with certain the Bombay phenotype.
pathologic conditions. In contrast, anti-H in non-Bombay individuals is usually IgM and
o Decreased levels of isoagglutinins may be seen in patients clinically insignificant.
with acquired and congenital hypogammaglobulinemia and Anti-IH is not uncommonly found in patient sera and is usually IgM;
agammaglobulinemia. compatible blood is easily found among donors of identical ABO
o Some individuals with roundworm infections will have type.
elevated levels of anti-A.

#MLSWATER2022 Page 5 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
INHERITANCE OF THE ABO BLOOD GROUPS o The specific combination of these four components
The theory for the inheritance of the ABO blood groups was first determines an individual's type in most cases.
described by Bernstein in 1924. The table below shows the possible permutations of antigens and
o He demonstrated that an individual inherits one ABO gene antibodies with the corresponding ABO type ("yes" indicates the
from each parent and that these two genes determine presence of a component and "no" indicates its absence in the
which ABO antigens are present on the RBC membrane. blood of an individual).
The inheritance of ABO genes, therefore, follows simple Mendelian
genetics. ABO Blood Antigen A Antigen B Antibody Antibody
o ABO, like most other blood group systems, is codominant in Type anti-A anti-B
expression. A YES NO NO YES
o One position, or locus, on each chromosome 9 is occupied B NO YES YES NO
by an A, B, or O gene. O NO NO YES YES
The O gene is considered an amorph, as no detectable antigen is AB YES YES NO NO
produced in response to the inheritance of this gene.
o Therefore, the group O phenotype is an autosomal For example;
recessive trait with the inheritance of two O genes that are People with type A blood will have the A antigen on the surface of
nonfunctional. their red cells (as shown in the table below).
o The designations group A and B refer to phenotypes, o As a result, anti-A antibodies will not be produced by them
whereas AA, BO, and OO denote genotypes. because they would cause the destruction of their own
o In the case of an O individual, both phenotype and blood.
genotype are the same, because that individual would have However, if B type blood is injected into their systems, anti-B
to be homozygous for the O gene. antibodies in their plasma will recognize it as alien and burst or
o An individual who has the phenotype A (or B) can have the agglutinate the introduced red cells in order to cleanse the blood of
genotype AA or AO (or BB or BO). alien protein.

THE ABO GENOTYPES AND PHENOTYPES. ABO Blood Antigen A Antigen B Antibody Antibody
Serologically, it is not possible to determine the genotype from the Type anti-A anti-B
phenotype of an A or B individual. A YES NO NO YES
o Family studies or molecular assays would have to be B NO YES YES NO
performed to determine the exact genotype. O NO NO YES YES
o The phenotype and genotype are the same in an AB AB YES YES NO NO
individual because of the inheritance of both the A and B
gene. Individuals with type O blood do not produce ABO antigens.
All humans and many other primates can be typed for the ABO Therefore, their blood normally will not be rejected when it is given
blood group. to others with different ABO types.
o There are four principal types: A, B, AB, and O. o As a result, type O people are universal donors for
o There are two antigens and two antibodies that are mostly transfusions, but they can receive only type O blood
responsible for the ABO types. themselves.

#MLSWATER2022 Page 6 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
Those who have type AB blood do not make any ABO antibodies. H AND SE ANTIGEN
Their blood does not discriminate against any other ABO type. The H antigen is actually the precursor structure on which A and B
o Consequently, they are universal receivers for transfusions, antigens are made.
but their blood will be agglutinated when given to people o Inheritance of the H gene results in the formation of the H
with every other type because they produce both kinds of antigen.
antigens. o The H and Se genes are closely linked and located on
chromosome 19, in contrast to ABO genes, which are
ABO Blood Antigen A Antigen B Antibody Antibody located on chromosome 9.
Type anti-A anti-B o The H and Se genes are not part of the ABO system;
A YES NO NO YES however, their inheritance does influence A and B antigen
B NO YES YES NO expression.
O NO NO YES YES The H gene must be inherited to form the ABO antigens on the
AB YES YES NO NO RBCs, and the Se gene must be inherited to form the ABO antigens
in secretions.
It is easy and inexpensive to determine an individual's ABO type from a
few drops of blood. PRECURSOR SUBSTANCE
o A serum containing anti-A antibodies is mixed with some of the The precursor substance on erythrocytes is referred to as type 2.
blood. o This means that the terminal galactose on the precursor
o Another serum with anti-B antibodies is mixed with the remaining substance is attached to the N-acetylglucosamine in a beta
sample. 1 → 4 linkage .
Whether or not agglutination occurs in either sample indicates the ABO
A type 1 precursor substance refers to a beta 1 → 3 linkage
type.
It is a simple process of elimination of the possibilities. between galactose and N-acetylglucosamine.
ABH antigens on the RBC are constructed on oligosaccharide chains
For instance, if an individual's blood sample is agglutinated by the anti-A
of a type 2 precursor substance.
antibody, but not the anti-B antibody, it means that the A antigen is
present but not the B antigen. Therefore, the blood type is A.
ABH ANTIGEN DEVELOPMENT
FORMATION OF ABH ANTIGENS ON THE RBCS FROM PRECURSOR The ABH antigens develop early in fetal life but do not increase
much in strength during the gestational period.
SUBSTANCE TO IMMUNODOMINANT SUGARS
The RBCs of the newborn have been estimated to carry anywhere
The formation of ABH antigens results from the interaction of genes from 25% to 50% of the number of antigenic sites found on the
at three separate loci (ABO, Hh, and Se). adult RBC.
o These genes do not actually code for the production of As a result, reactions of newborn RBCs with ABO reagent antisera
antigens but rather produce specific glycosyltransferases are frequently weaker than reactions with adult cells.
that add sugars to a basic precursor substance. The expression of A and B antigens on the RBCs is fully developed by
A, B, and H antigens are formed from the same basic precursor 2 to 4 years of age and remains constant throughout life.
material (called a paragloboside or glycan) to which sugars are In addition to age, the phenotypic expression of ABH antigens may
attached in response to specific enzyme transferases elicited by an vary with race, genetic interaction, and disease states.
inherited gene.

#MLSWATER2022 Page 7 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
INTERACTION OF Hh AND ABO GENES BLOOD GROUP A
In the formation of blood group A, the A gene (AA or AO) codes for
Blood Group O
the production of α-3-N-acetylgalac - tosaminyltransferase, which
Individuals who are blood group O inherit at least one H gene
transfers an N-acetyl-D-galac - tosamine(GalNAc) sugar to the H
(genotype HH or Hh) and two O genes.
substance.
The H gene elicits the production of an enzyme called α-2-L-
o This sugar is responsible for A specificity (blood group A).
fucosyltransferase, which transfers the sugar L-fucose to an
o The A-specific immunodominant sugar is linked to a type 2
oligosaccharide chain on the terminal galactose of type 2 chains.
precursor substance that now contains H substance through
o The sugars that occupy the terminal positions of this
the action of the H gene.
precursor chain and confer blood group specificity are called
The A gene tends to elicit higher concentrations of transferase than
the immunodominant sugars.
the B gene. This leads to the conversion of practically all of the H
o Therefore, L-fucose is the sugar responsible for H specificity
antigen on the RBC to A antigen sites.
(blood group O).
o As many as 810,000 to 1,170,000 antigen sites exist on an
The O gene at the ABO locus, which is sometimes referred to as an
A1 adult RBC in response to inherited genes.
amorph, does not elicit the production of a catalytically active
polypeptide transferase; therefore, the H substance remains
unmodified. BLOOD GROUP B
Consequently, the O blood group has the highest concentration of Individuals who are blood group B inherit a B gene (BB or BO) that
H antigen. codes for the production of α-3-D-galactosyltransferase, which
o The H substance (L-fucose) must be formed for the other attaches D-galactose (Gal) sugar to the H substance previously
sugars to be attached in response to an inherited A and/or B placed on the type 2 precursor substance through the action of the
gene. H gene.
The H gene is present in more than 99.99% of the random o This sugar is responsible for B specificity (blood group B).
population. Its allele, “h,” is quite rare, and the genotype hh is o Anywhere from 610,000 to 830,000 B antigen sites exist on
extremely rare. a B adult RBC in response to the conversion of the H antigen
The term Bombay has been used to refer to the phenotype that by the α-3-Dgalactosyltransferase produced by the B gene.
lacks normal expression of the ABH antigens because of the When both A and B genes are inherited, the B enzyme (α-3-D-
inheritance of the hh genotype. galactosyltransferase) seems to compete more efficiently for the H
o The hh genotype does not elicit the production of α-2-L- substance than the A enzyme (α-3-
fucosyltransferase. Nacetylgalactosaminyltransferase).
o Therefore, L-fucose is not added to the type 2 chain, and H o Therefore, the average number of A antigens on an AB adult
substance is not expressed on the RBC. cell is approximately 600,000 sites, compared with an
o Even though Bombay (hh) individuals may inherit ABO average of 720,000 B antigen sites.
genes, normal expression, as reflected in the formation of A,
B, or H antigens, does not occur.

#MLSWATER2022 Page 8 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
REACTIVITY OF ULEX EUROPAEUS WITH THE VARIOUS ABO GROUPS NOTE
The H antigen is a basic blood group antigen present in human beings. 1. In vitro diagnostic reagent for laboratory and professional use only.
There is considerable variation in the H antigen content in different  Not for medicinal use.
individuals of the same ABO group but the general pattern indicates 2. The reagent contains sodium azide 0.1% as preservative. Avoid
their strength as O>A2>A2B>B>A1>A1B. contact with skin and mucosa.
Water soluble H substance can also be demonstrated in saliva or body  On disposal flush with large quantities of water.
fluids of individuals who are secretors. 3. Extreme turbidity may indicate microbial contamination / reagent
deterioration. Such reagents should be discarded.
Human red blood cells that do not agglutinate with Anti-H lectin are
classified as Bombay Phenotype (Oh).
ABO SUBGROUPS
o The Bombay Phenotype is more common in India than other parts
of the world and the estimated gene frequency of Oh phenotype in A AND B SUBGROUPS
Bombay is 0.0066%. A1 AND A2 SUBGROUPS
Group A antigens can be differentiated into multiple subgroups. The
REAGENT two major subgroups are
o A1, 80% of Group A individuals, and
Anti-H lectin is a ready to use purified extract of Ulex europaeus seeds.
o A2, 20% of group A individuals
It contains a phytohaemagglutinin, which is virtually specific for the H
antigen on human red blood cells. Persons typing as AB can be divided into the same percentages of A
antigen presentation.
Anti-H lectin is used for recognition of the H antigen on human red
o A1B make up approximately 80% and
blood cells.
o A2B are 20% of all AB individuals.
It is useful, especially for assessing the H secretor status of group 'O'
The remaining group A individuals fall into one of many minor
individuals and also in differential grouping of Aint subgroup along with
subgroups.
Anti-A1 lectin.

A1 and A2 antigens have qualitative and quantitative differences.


PRINCIPLE
Qualitative differences of A1 and A2 antigens
Human red blood cells possessing the H antigen will agglutinate in
the presence of seed extract (lectins) containing For qualitative test, a subgroup phenotype are distinguished from A1
phytohaemagglutinin specifically directed towards it. phenotype by a glycolipid antigen expression because of the
Water soluble H substance present in saliva neutralises Anti-H structural differences in the branching of the oligosaccharide chain
lectin. The structural difference explain why the A subgroup individuals
Agglutination of red blood cells / Neutralization of Anti-H lectin by often make the anti-A1
saliva is a positive test result and indicates the presence of The reagent dolichos biflorus lectin will distinguish A1 from A2
H substance on/in the red cell / saliva respectively. When red cells are qualitatively tested for antigens, A1 and A2 red
No agglutination / Neutralization of Anti-H lectin is a negative test cells have differing amounts of antigens on the cell surface.
result and indicates the absence of H substance on / in the red cell / o The A1 gene produces a transferase that has a greater ability to
saliva respectively. convert H antigen to A antigen than the A2 gene.
o This quantitative difference results from the kinetics of the
reaction catalyzed by each of the transferases.
There are also differences in the quantity of transferase produced.

#MLSWATER2022 Page 9 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
o Individuals exhibiting the A1 phenotype have five to ten times QUANTITATIVE QUALITATIVE
more transferase than those with an A2 phenotype. ↓ Number of Antigen Sites Differences in the precursor
oligosaccharide chains
Two mutations have been detected that produce this A2 phenotype: ↓ Amount of Transferase Enzyme Subtle differences in transferase
o a Pro156Leu substitution enzymes
o a single nucleotide deletion (nucleotide 1060). ↓ Amount of Branching Formation of anti-A1, in a
These substitutions are responsible for the decreased enzyme activity percentage of some subgroups
that differentiates A2 from A1 cells. -Refer to Module packet regarding Quali & Quanti differences of A1 &A 2 antigens
(pp. 11-12)
The antigens also differ qualitatively.
ADDITIONAL A SUBGROUPS
Quantitative differences of A1 and A2 antigens
Occurring subgroups of A exist less frequently.
For quantitative test, produces so little antigen because the number
o These subgroups are also genetically controlled.
of A antigen is reduced in A2.
Subgroups of A include Aintr, A3, Ax , Am, Aend, Ael,and Abantu.
There is a little amount of antigen in A2 than in A1
o These subgroups follow the patterns of A1 and A2 with regard to
A2 antigens are composed mainly of linear oligosaccharide chains
quantitative and qualitative antigenic differences.
A1 cells have a greater number of branched chains o The cells of these subgroups exhibit fewer antigen sites on their
o In routine testing, this qualitative difference is not detectable but surface while many demonstrate an anti-A1 in the plasma.
can be determined biochemically.
Adsorption and elution techniques may be necessary for detection of
Typing of A1 and A2 cells is unremarkable with routine antisera. antigens on the surface of red cells.
Both A1 and A2 cells will react equally with anti-A and anti-A,B. The classification of subgroups is based on reactions of the patient’s
The lectin, Dolichos biflorus, is used to obtain an extract with anti-A1 red cells with anti-A, anti-B, anti-A,B, and anti-A1 antisera as well as A1,
specificity. A2, and B reverse grouping cells.
o Dolichos biflorus will react specifically with A1 cells and will be o While testing for subgroups of A, a mixed field agglutination
negative with A2 cells. reaction may be noted.
o A3 cells will demonstrate this pattern of agglutination with anti-A
A2 individuals can develop antibodies to the A1 antigens. and anti-A,B.
The typical reaction pattern of reverse grouping in a group A individual
is no agglutination with the A cells (no anti-A) and agglutination with B SUBGROUPS OF B
cells (anti-B present). Very rare and encountered less frequently than subgroups of A.
o In A2 persons with an anti-A1, the A cells will also be agglutinated in Methods of detection and classification are similar to those used for the
the reverse grouping. subgroups of A.
o This discrepancy should be confirmed by testing the red cells with
the Dolichos biflorus lectin. WEAK SUBGROUPS OF A
The prevalence of A subgroups of A weaker than A1 and A2 is less than
1%.
Fewer antigen sites on the RBC means weaker reactions with antisera.
It is possible for an Ax donor to be mistyped as O.

#MLSWATER2022 Page 10 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
This unit could then be transfused into an O recipient, who has anti- THE BOMBAY PHENOTYPES (OH)
A,B. The Bombay phenotype was first reported by Bhende in 1952 in
The anti-A,B antibody in the recipient could agglutinate and lyse the Bombay, India.
donor Ax RBCs and cause intravascular hemolysis. It represents the inheritance of a double dose of the h gene,
producing the very rare genotype hh.
Subgroup Laboratory Results Number of A o As a result, the ABO genes cannot be expressed, and ABH
Antigens sites antigens cannot be formed, since there is no H antigen
A3 Mixed field reaction with anti-A and 35,000 per made in the Bombay phenotype.
most anti-A,B reagents RBC More than 130 Bombay phenotypes have been reported in various
Ax Characteristically not agglutinated 4000 parts of the world. These RBCs are devoid of normal ABH antigens
with anti-A but do agglutinate with most and, therefore, fail to react with anti-A, anti-B, and anti-H.
examples of anti-A,B In RBC testing using anti-A and anti-B, the Bombay would
Aend Mixed field reaction with anti-A and 3500 phenotype as an O blood group.
anti-A,B. Aend is inherited as an allele at Using conventional or non-potent anti-A and anti-B sera
the ABO locus. Anti-A1 is found in some would result in the Bombay phenotype to be mistaken as
sera. Only H is found in secretions. O-blood group.
Am Characteristically not agglutinated, or very 200-1900 To differentiate group O from Bombay, use anti-A, anti-B,
weakly agglutinated by all anti-A and anti- and anti-H since group O (with H-antigens) would always
A,B have an agglutination with anti-H
reagents. Usually do NOT produce anti-A1 o However, the RBCs of the Bombay phenotype (Oh) do not
in react with the anti-H lectin (Ulex europaeus), unlike those of
sera. the normal group O individual, which react strongly with
anti-H lectin.
WEAK B SUBGROUPS Bombay serum contains antiA, anti-B, anti-A,B, and anti-H.
With the addition of the anti-A and anti-B reagent to the cell, there Unlike the anti-H found occasionally in the serum of A1 and A1B
can be two reactions: weak agglutination or no agglutination at individuals, the Bombay anti-H can often be potent and reacts
all. strongly at 37°C.
It is an IgM antibody that can bind complement and cause RBC
Weak
lysis.
B(3) – mix field reaction with anti-A and anti-B
Transfusing normal group O blood (with the highest concentration
B(x)– weak agglutination with anti-A and anti-AB only
of H antigen) to a Bombay recipient (anti-H in the serum) would
cause immediate cell lysis.
No agglutination o Therefore, only blood from another Bombay individual will
B(m) – demonstrates the presence of A-substance be compatible and can be transfused to a Bombay recipient.
B(el) – shows the presence of H-substance ABH substance is also absent in saliva.
The (Oh) Bombay phenotype is inherited as an autosomal recessive
-Refer to reference book for further information. trait.
The underlying molecular defect is most commonly a mutation in
the gene FUT1 (H gene), which produces a silenced gene that is

#MLSWATER2022 Page 11 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
incapable of coding for the enzyme, α(1,2)fucosyltransferase (H Hodgkin’s disease also has been reported to weaken or depress
transferase). ABH red cell antigens, resulting in variable reactions during forward
This enzyme catalyzes the transfer of fucose in an α-1,2 linkage to grouping similar to those found in leukemia.
the terminal galactose of the precursor molecule on RBCs forming o The weakening of the antigen tends to follow the course of
the H antigen. the disease.
This mutation underlying the Bombay phenotype is also associated o The antigen strength will increase again as the patient
with a silenced FUT2 gene (Se gene), which normally encodes a very enters remission.
similar α(1,2)fucosyltransferase and normally transfers a fucose to The isoagglutinins (anti-A, anti-B, or anti-A,B) also may be weak or
form H antigens in secretions when active. absent in those leukemias demonstrating
When family studies demonstrate which ABO genes are inherited in hypogammaglobulinemia, such as chronic lymphocytic leukemia
the Bombay phenotype, the genes are written as superscripts (OhA, (CLL).
OhB, OhAB). Various lymphomas, such as the malignant (non-Hodgkin’s) variety,
may yield weak isoagglutinins, owing to moderate decreases in the
EFFECTS OF DISEASE ON THE EXPRESSION OF ABH ANTIGENS gamma globulin fraction.
AND ANTIBODIES Also, immunodeficiency diseases, such as congenital
ABH ANTIGENS AND ANTIBODIES IN DISEASE agammaglobulinemia, will also yield weak or absent isoagglutinins.
Associations between ABH antigens and practically any disorder o If this problem is suspected, a simple serum protein
known to man can be found throughout medical literature. electrophoresis will confirm or rule out this condition.
Even more profound are the associations of blood group specificity Individuals with intestinal obstruction, carcinoma of the colon or
with such things as a more pronounced: rectum, or other disorders of the lower intestinal tract may have
o “hangover” in A blood groups increased permeability of the intestinal wall, which allows passage
o “criminality” in group B blood groups of the bacterial polysaccharides from Escherichia coli serotype O86
o “good teeth” in group O individuals. into the patient’s circulation.
There are also several papers correlating blood groups with o This results in the acquired B phenomenon in group A1
personality traits. It is no surprise that many scientists refer to individuals.
these associations as a part of blood group mythology. o The patient’s group A red cells absorb the B-like
However, more relevant associations between blood groups and polysaccharide, which reacts with human-source anti-B.
disease are important to the blood banker in terms of blood group A lack of detectable ABO antigens can occur in patients with
serology. carcinoma of the stomach or pancreas.
Various disease states seem to alter red cell antigens and result in o The patient’s red cell antigens have not been changed, but
progressively weaker reactions or additional acquired the serum contains excessive amounts of blood group–
pseudoantigens, which can be seen during forward grouping. specific soluble substances (BGSS) that may neutralize the
Leukemia, chromosome 9 translocations, and any hemolytic antisera utilized in the forward grouping.
disease that induces stress hematopoiesis (e.g., thalassemia) have All these disease states previously mentioned may result in
been shown to depress antigen strength. discrepancies between the forward and reverse groupings,
o Often the cells will appear to show a mixed-field indicating that the patient’s red cell group is not what it seems.
agglutination (tiny agglutinates in a sea of unagglutinated All ABO discrepancies must be resolved before blood for transfusion
cells). is released for that patient.
o In some cases, secretor or molecular studies may help
confirm the patient’s true ABO group.
#MLSWATER2022 Page 12 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
LABORATORY TESTS TO DETECT AND IDENTIFY ABO Routine testing for antigens and antibodies is performed as a
ANTIGENS AND ANTIBODIES forward and reverse grouping, respectively.
ENZYME-CONVERTED O CELLS (ECO) The forward grouping is a test performed for antigens using known
antisera with patient’s cells that may contain unknown antigens.
Blood group O is considered the universal donor because it can be
Test methods for forward grouping include:
transfused to patients of all ABO types. Therefore, enzymes that
o Tube typing
remove terminal carbohydrates from the nonreducing end of
o Gel Technology
carbohydrate chains could be used to remove terminal A and B
o Automation
sugars to convert the blood supply to all universal group O units.
o Solid Phase Technology
An enzyme from coffee beans, α-galactosidase, has been the most
ABO forward grouping with tube typing uses a saline suspension of
successful at removing galactose to convert blood group B to group
3 to 5% washed patient red cells. These cells are combined in a 1:1
O.
ratio with commercial antisera.
RBCs treated in this manner have normal survival when transfused
When utilizing gel technology, the cell suspension consists of a
to group B, A, or O recipients.
0.8% suspension of washed patient cells in the manufacturer’s
Removal of N-acetylgalactosamine to convert group A to group O
recommended diluent.
has been much more problematic, owing to the inaccessibility of the
These cells are applied to the anti-A and anti-B tubes of the gel card.
carbohydrates on internal branching chains, especially those found
In both methods, centrifugation is applied and results interpreted.
on A1 cells.
The procedures required to convert B to O, which include exposure
to low pH followed by numerous washings, make them impractical ANTISERA
for general use. This is an active area of research and alternative When performing tube typing, three antiseras are available for ABO
enzymes and improved methodologies are under investigation. forward grouping.
Forward grouping may be performed using all three, or in the case
BLOOD TYPING of patients or transfusion recipients anti-A and anti-B are used.
Antisera are combined in a 1:1 ratio with the patient’s cell
Blood typing is a method to tell what type of blood you have.
suspension.
Is done so you can safely donate your blood or receive a blood
When evaluating reaction patterns, the antigens on the cells are
transfusion.
reacting with the specific antibodies in the antisera.
It is also done to see if you have a substance called Rh factor on the
o Group A individual has the A antigen and reacts with both
surface of your red blood cells.
anti-A and anti-A,B.
Your blood type is based on whether or not certain proteins are on
o A group O will react with no antisera since these cells have
your red blood cells.
neither A nor B antigens.
o These proteins are called antigens. Your blood type (or
The inclusion of anti-A,B antisera in forward grouping is significant.
blood group) depends on what types your parents passed
o It is not a mixture of anti-A and anti-B, but rather a separate
down to you.
antibody that will react with both the A and B antigens.
It is included in forward grouping and serves two purposes:
FORWARD AND REVERSE GROUPING 1. To confirm the results of the anti-A and anti-B.
ABO FORWARD GROUPING 2. To detect weak subgroups of A and B.
As previously described, ABO antigens are present on the surface of These subgroups may demonstrate a positive reaction with anti-A,B
red cells, while the antibodies are found in plasma or serum. but not with anti-A and anti-B.

#MLSWATER2022 Page 13 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
SAMPLE PROCEDURE 1: REACTION PATTERNS FOR ABO GROUP
ABO FORWARD GROUPING: TUBE TYPING METHOD BLOOD GROUP ANTI-A ANTI-B ANTI-A,B
Procedure: A POSITIVE NEGATIVE POSITIVE
1. Prepare a 3 to 5% suspension of patient’s red cells. B NEGATIVE POSITIVE POSITIVE
2. Label three small test tubes with the patient’s name and AB POSITIVE POSITIVE POSITIVE
identification number. O NEGATIVE NEGATIVE NEGATIVE
3. Each of these tubes should then be labeled as follows: First tube: In Forwarding Typing the Antigens are used as Antisera.
“Anti-A” Second tube: “Anti-B” Third tube: “Anti-A, B” o Blood Type O has no agglutination
NOTE: Labeling should be done with care since clerical errors are the o Blood Type A only has the reaction seen in Anti-A since Blood type A
most frequent errors in the blood bank. contains Antigen A
4. Check clarity and expiration date on antisera; record information. o Blood Type B only has the reaction seen in Anti-B since Blood type B
5. To each of these tubes, add one drop of the corresponding antisera. contains Antigen B
NOTE: Use a free-floating drop. Do not touch the dropper to the side o Blood Type AB has both the Antigen AB therefore it is present in
of the tube. Always add antisera before cells. both reactions.
6. Using a transfer pipet, add one drop of the well-mixed 5% cell
suspension to each of these three tubes. Molecular Testing
NOTE: Use a free-floating drop. Do not touch the pipet to the side of
As molecular diagnostic testing continues to develop, applications
the tube.
to ABO forward grouping may become commonplace in the
7. Gently mix all tubes.
laboratory.
8. Serofuge all three test tubes for 15 seconds.
Molecular testing has the potential to solve typing discrepancy in
NOTE: Time may vary with each serofuge. Check the calibration
recently or chronically transfused patients.
information for each individual serofuge.
These patients present unique challenges to the blood bank, since
9. Remove each tube and examine for hemolysis.
typing through traditional methods frequently produces discrepant
10. Using an agglutination viewer, gently resuspend each cell button,
or erroneous results.
and examine for agglutination.
11. Grade each reaction and record the results. POLYMERASE CHAIN REACTION (PCR) METHODS
Have been proven to be more reliable than traditional serological
methods in resolution of typing discrepancies in recipients that have
SAMPLE PROCEDURE 2:
been transfused within the last 30 days.
1. Prepare a 0.8% suspension of test cells in the appropriate diluent.
As PCR test results are available within several hours, this testing
2. Choose a gel card for ABO forward grouping and label with patient
method presents a promising future for resolution of discrepancies
identification.
that were previously major compatibility challenges.
3. Add 10μl of cell suspension of test cells to each microtube on the
card.
4. Centrifuge the card. Reverse Grouping
5. Evaluate each microtube for agglutination. Record and interpret
ABO reverse grouping uses patient plasma combined in a 2:1 ratio
results.
with commercially prepared cells.
The cells are packaged in sets of two (A1 and B) or three (A1, A2,
and B). The cells are used to detect unknown antibodies in the
plasma.

#MLSWATER2022 Page 14 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
The result is evaluated by examining the tubes for hemolysis and INTERPRETATION OF REVERSE GROUPING TEST RESULTS
agglutination. BLOOD GROUP A1 CELLS B CELLS
NOTE: A NEGATIVE POSITIVE
Recall that antibodies present in the test plasma correspond to B POSITIVE NEGATIVE
antigens missing on the red cell surface. O POSITIVE POSITIVE
For example, group A has the A antigen and the B antigen is not AB NEGATIVE NEGATIVE
present. For the reverse typing, this is the detection of antibodies in our serum
The corresponding B antibody is demonstrated in the group A o Both positive reaction for Blood type O since it contains both Anti
individual’s plasma. A,B
When the plasma reacts with the reagent red blood cells, the B o In Blood type A, only B cells has the reaction since Blood type A
antibody reacts with specific antigens on the B cells, but not individual has the Anti B
antigens on the A cells. o In Blood type B, only A cells has the reaction since Blood type B has
Therefore, a positive reaction will be seen in the B tube, but not in the Anti- A
the A tube. o In Blood type AB, there is no reaction for the reverse typing since
Blood type AB contains no any antibodies in their serum
SAMPLE PROCEDURE:
1. Label two 10 × 75 test tubes with the patient’s name and
identification number. ABO DISCREPANCIES
2. Label one of the tubes A and one of them B. ABO discrepancies occur when unexpected reactions occur in the
NOTE: Labeling is a crucial step in the blood typing procedure. Fatal forward and reverse grouping.
errors are made when clerical errors occur. These can be due to:
3. Add two drops of serum to each tube. o Problems with the patient’s serum (reverse grouping)
4. To the appropriate tube, add one drop of well-mixed reagent red o Problems with the patient’s red cells (forward grouping)
cells. o Problems with both the serum and cells.
NOTE: Before adding reagent red cells, be certain they are well- The unexpected reaction can be due to an extra positive reaction or
mixed and that all of the cells are resuspended from the bottom of a weak or missing reaction in the forward and reverse grouping.
the vial. All ABO discrepancies must be resolved prior to reporting a patient
5. Gently mix the two tubes. or donor ABO group.
6. Serofuge for 15 seconds.
NOTE: The time for centrifugation may vary with each serofuge. Technical Errors
Check the calibration on the serofuge being used for testing. Technical errors can also cause ABO discrepancies.
7. Remove each tube and examine for hemolysis. This includes errors in labeling the blood sample at the patient’s
8. Using an agglutination viewer, gently resuspend each cell button and bedside or in the laboratory; therefore, patient and sample
examine for agglutination. identification are essential!
9. Grade and record each reaction. Other errors include the failure to add reagents or the addition of
incorrect reagents or sample. Therefore, it is recommended that
serum and antiserum be added first, then the patient or reagent red
cells.

#MLSWATER2022 Page 15 of 16
MODULE 2 PART 2- ABO BLOOD GROUP SYSTEM
It is also recommended that results be recorded immediately to
Forward vs. Reverse Typing
avoid transcription errors.
In addition, contaminated reagents can cause errors in testing. Forward Typing Reverse Typing
Therefore, looking at all the reagent vials when performing ABO Other term Direct or Front Typing Indirect or Back Typing
testing and during quality control testing is extremely important. Detection A & B antigen within Anti-A and Anti B in
patient’s RBCs patient’s serum
Resolution Specimen 1 drop of whole blood or 2- 2 drops of patient’s serum
5% red cell suspension
If the initial test was performed using RBCs suspended in serum or
Reagent 2 drops of Anti-A (blue)and 1 drop of A1 red cells and B
plasma, repeat testing the same sample using a saline suspension of
Anti-B (yellow) red cells derived from
RBCs can usually resolve the ABO discrepancy.
human source, 4-5% RBC
It is important to make sure that any and all technical factors that
suspension is used as
may have given rise to the ABO discrepancy are reviewed and
reagents
corrected.
It is also essential to acquire information regarding the patient’s Blood Forward Typing Antigens Reverse Typing Antibody
age, diagnosis, transfusion history, medications, and history of Group Px’s RBC with in RBCs Px’s Serum with in Serum
pregnancy. Anti-A Anti-B A1 Cells B Cells
If the discrepancy persists and appears to be due to an error in
O 0 0 No A/ B + + Anti-A &
specimen collection or identification, a new sample should be
Anti-B
drawn from the patient and the RBC and serum testing repeated.
A + 0 A Ag 0 + Anti-B
When a discrepancy is encountered, results must be recorded, but
B 0 + B Ag + 0 Anti-A
interpretation of the ABO type must be delayed until the
AB + + A&B 0 0 none
discrepancy is resolved.
Px: Patient; 0: Negative/ No reaction; +: Positive
If blood is from a potential transfusion recipient, it may be
necessary to administer group O–compatible RBCs before the
discrepancy is resolved. Investigation of Weak A Subgroup: https://drive.google.com/file/d/1z-
In general, when investigating ABO discrepancies, it should be 4kYqsCKm7Yz2oMklMuV9FiixmEylrN/view?usp=sharing
noted that RBC and serum grouping reactions are very strong (3+ to
4+); therefore, the weaker reactions usually represent the Investigation of Weak B Subgroup:
discrepancy. https://drive.google.com/file/d/16C6F2j8rxVVBdKVw8NkPSiT6QHJ34Tn6/vi
ew?usp=sharing
Categories of ABO Discrepancies
-END OF TRANSCRIPTION -
ABO discrepancies may be arbitrarily divided into four major Bloody hell isn’t it? But imagine the lives that you will
categories: group I, group II, group III, and group IV discrepancies. save in the future!
Be Positive (B+) Dreamer!
-Refer to ABO Discrepancies Assignment for the complete description of each
Category. -quote by no one.

APPENDICES
#MLSWATER2022 Page 16 of 16
MODULE 2 Part 3: Rh-Hr Blood Group System
MLS 18, March 10, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
HISTORY OF THE RH SYSTEM
Legend: Before 1939, the only significant blood group antigens recognized were
Notes/Module Packet Bullet those of the ABO system.
MODULE OUTLINE Transfusion medicine was thus based on matching ABO groups.
I. History of the Rh System
II. Nomenclatures of the Rh System Levine and Stetson
A. Fisher Race: DCE Terminology Described a hemolytic transfusion reaction in an obstetrical patient.
B. Wiener: Rh-Hr Terminology o After delivery of a stillborn infant, a woman required transfusions.
C. Rosenfield and Coworkers: Alphanumeric Terminology Her husband, who had the same ABO type, was selected as her
D. ISBT Committee: Numeric Terminology donor.
III. Proposed Genetic Pathways o After transfusion, the recipient demonstrated the classic symptoms
A. Mechanisms of Antigen Production of an acute hemolytic transfusion reaction.
B. Biochemistry of the Rh Antigens Subsequently, an antibody was isolated from the mother’s serum that
IV. Weak D: Variations of the Rho (D) Antigen Expression reacted both at 37C and at 20C with the father’s RBCs.
V. Detection of Rh Antibodies and Antigens It was postulated that the fetus and the father possessed a common
VI. Clinical Consideration factor that the mother lacked.
VII. Rh Deficiency Syndrome: Rhnull and Rhmod o While the mother carried the fetus, she was exposed to this factor
VIII. Unusual Phenotypes and Rare Alleles and subsequently built an antibody that reacted against the
IX. The LW Antigen transfused RBCs from the father, which resulted in the hemolytic
transfusion reaction.

LEARNING OUTCOMES Landsteiner and Wiener


1. Explain the derivation of the term Rh. Reported on an antibody made by guinea pigs and rabbits when they
2. Differentiate Rh from LW blood group systems were transfused with rhesus monkey RBCs a year after Levine and
3. Compare and contrast the Fisher-Race and Weiner theories of Rh Stetson’s discovery.
inheritance.
This antibody, which agglutinated 85 percent of human RBCs, was
4. Differentiate the mechanisms that result in the weakened
named Rh.
expression of D on the red blood cells.
Demonstrated that the agglutinin that had caused the hemolytic
5. List some instances in which the weak-D status of an individual
transfusion reaction and the antibody described by Landsteiner and
must be determined.
Wiener appeared to define the same blood group.
6. Describe the characteristics of Rh antigens (excluding dCcEe).
o Many years later it was recognized that the two antibodies were
7. Describe the characteristics of Rh antibodies.
different.
8. Compare & contrast Rhnull&Rhmod
o However, the name Rh was retained for the human-produced
9. Describe the role of RhAG in Rh antigen expression.
antibody, and the anti-rhesus antibody formed by the animals was

#MLSWATER2022 Page 1 of 9
MODULE 2 Part 3: Rh-Hr Blood Group System
renamed anti-LW in honor of those first reporting it (Landsteiner dictates one’s phenotype (the antigens expressed on the RBC that
and Wiener). can be detected serologically).
Further research resulted in defining Rh as a primary cause of hemolytic An individual’s Rh phenotype is reported as DCE rather than CDE
disease of the newborn (HDN, also called erythroblastosis fetalis) and a because Fisher postulated that the C/c locus lies between D/d and E/e
significant cause of hemolytic transfusion reactions. loci. This information is based on frequencies of the various gene
By the mid-1940s, five antigens made up the Rh system. Today the Rh combinations.
blood group system is made up of nearly 50 different specificities. Rh-positive person exhibiting a deletion phenotype such as these is
written -De or -DE, CD- or cD-, or -D-, respectively.
o The last is sometimes referred to as a double deletion.
NOMENCLATURES OF THE RH SYSTEM o The person expressing no Rh antigens on the RBC is said to be
The terminologies used to describe the Rh system are derived from four Rhnull, and the phenotype may be written as –––/–––.
sets of investigators: Weakened expression of all Rh antigens of an individual has also been
o Two of the terminologies are based on the postulated genetic reported.
mechanisms of the Rh system. o These individuals are said to have the Rhmod phenotype, and there
o The third terminology describes only the presence or absence of a is no unique way of indicating this using the Fisher-Race terminology
given antigen.
o The fourth is the result of the combined efforts of the International
Society of Blood Transfusion (ISBT) Working Party on Terminology
for Red Cell Surface Antigens.

Fisher-Race: The DCE Terminology


In the early 1940s, Fisher and Race were investigating the antigens
found on human RBCs, including the newly defined Rh antigen.
o They postulated that the antigens of the system were produced by
three closely linked sets of alleles.
Each gene was responsible for producing a product (or antigen) on the
RBC surface. Each antigen and corresponding gene were given the same
letter designation (when referring to the gene, the letter is italicized).
Fisher and Race named the antigens of the system D, d, C, c, and E and
e.
o Now it is known that “d” represents the absence of D antigen.
Wiener: The Rh-Hr Terminology
o The phenotype (blood type observed during testing) of a given RBC
is defined by the presence of D, C, c, E, and e expression. Wiener believed that the gene responsible for defining Rh actually
According to the Fisher-Race proposal, each person inherits a set of Rh produced an agglutinogen that contained a series of blood factors.
genes from each parent (i.e., one D or d, one C or c, and one E or e). According to Rh-Hr terminology, this Rh gene produces at least three
o Because Rh genes are codominant, each inherited gene expresses factors within an agglutinogen.
its corresponding antigen on the RBC. o The agglutinogen may be considered the phenotypic expression of
o The combination of maternal and paternal haplotypes determines the haplotype. Each factor is an antigen recognized by an antibody.
one’s genotype (the Rh genes inherited from each parent) and Antibodies can recognize single or multiple factors (antigens).
#MLSWATER2022 Page 2 of 9
MODULE 2 Part 3: Rh-Hr Blood Group System
Rosenfield and Coworkers: Alpha/ Numeric Terminology
As the Rh blood group system expanded, it became more difficult to
assign names to new antigens using existing terminologies.
In the early 1960s Rosenfield and associates proposed a system that
assigns a number to each antigen of the Rh system in order of its
discovery or recognized relationship to the Rh system.
o This system has no genetic basis, but simply demonstrates the
presence or absence of the antigen on the RBC.
o A minus sign preceding a number designates absence of the
antigen.
For the five major antigens, D is assigned Rh1, C is Rh2, E is Rh3, c is Rh4,
and e is Rh5.
For RBCs that type D C E c negative, e negative, the Rosenfield
designation is Rh: 1, 2, 3, 4, 5. If the sample was not tested for e, the
designation would be Rh: 1, 2, 3, 4. All Rh system antigens have been
When describing an agglutinogen, the uppercase R denotes the assigned a number.
presence of the original factor, the D antigen.
The lowercase r indicates the absence of the D antigen. International Society of Blood Transfusion: Numeric Terminology
The presence of uppercase C is indicated by a one (1) or a single
The ISBT adopted a six-digit number for each authenticated blood
prime (′). group specificity.
Lowercase c is implied when there is no 1 or ′ indicated. o The first three numbers represent the system and the remaining
three the antigenic specificity.
o The number 004 was assigned to the Rh blood group system, and
When referring to the Rh antigens (or factors) in Wiener nomenclature,
then each antigen assigned to the Rh system was given a unique
o the single prime (′) refers to either C or c, number to complete the six-digit computer number.
o The double prime (′′) to either E or e. o 001 was assigned to -------
o Therefore, D is RH1, C is RH2, and so forth.
o If the r precedes the h (i.e., rh′ or rh′′), this refers to the C or E
The phenotype designation includes the alphabetical symbol that
antigens, respectively. denotes the blood group, followed by a colon and then the specificity
o When the h precedes the r, this refers to either the c (hr′) or e (hr′′) numbers of the antigens defined.
antigen. A minus sign preceding the number indicates that the antigen was
Rh0 is equivalent to D. In the Wiener nomenclature, there is no tested for but was not present.
designation for the absence of D antigen. o The phenotype D CE c e or DccEe or R2 r would be written RH:1,-
2,3,4,5.
When referring to a gene, an allele, or a haplotype, the symbols are
italicized, followed by a space or asterisk, and then the numbers of the
specificities are separated by commas.
o R1 or DCe would be RH 1,2,5.

#MLSWATER2022 Page 3 of 9
MODULE 2 Part 3: Rh-Hr Blood Group System
PROPOSED GENETIC PATHWAYS WEAK D: VARIATIONS O F THE RH0 (D) ANTIGEN EXPRESSION
MECHANISMS OF ANTIGEN PRODUCTION RBCs carrying the weaker D antigen have historically been referred to as
Two theories of Rh genetic control were initially postulated. having the Du type.
o Wiener postulated that a single gene produces a single product that o Now they are referred to as expressing weak D and are considered
contains separately recognizable factors. Rh-positive.
o In contrast, Fisher and Race proposed that the Rh locus contains o Three different mechanisms have been described that can explain
three distinct genes that control the production of their respective the weakened expression of the D antigen:
antigens.
It is currently accepted that only two closely linked genes located on 1. Genetic Weak D
chromosome 1 control the expression of Rh; one gene (RHD) codes for Inheritance of D genes that code for a weakened expression of the D
the presence or absence of the D polypeptides and the second gene antigen.
(RHCE) for either RHCe, RHcE, RHce, or RHCE polypeptides. The D antigens expressed appear to be complete but few in number. On
Another gene (RHAG), which resides on chromosome 6, produces a Rh- a molecular level, these quantitative differences in D expression are
associated glycoprotein that is very similar in structure to the Rh attributed to mutations of the Rh polypeptide.
proteins and within the RBC membrane forms complexes with the Rh Inheritance of these genes can be tracked vertically from one
polypeptides. RHAG is called a coexpressor and must be present for the generation to the next and are seen most frequently in blacks. The
successful expression of the Rh antigens but by itself does not express genetic weak D is rare and seldom found in whites.
any of the Rh antigens.
The Rh genes are inherited as codominant alleles. Offspring inherit one 2. C Trans
Rh haplotype from each parent. In rare instances, individuals express no Position effect or gene interaction effect
Rh antigens on their RBCs. These individuals are said to have the Rhnull
In individuals showing the gene interaction weak D, the allele carrying D
phenotype.
is trans (or in the opposite haplotype) to the allele carrying C
o for example, Dce/dCe.
BIOCHEMISTRY OF THE RH ANTIGENS
The Rh antigen on the RBC is normal, but the steric arrangement of the
Rh antigens are nonglycosylated protein. C antigen in relationship to the D antigen appears to interfere with the
o This means that there are no carbohydrates attached to the protein. expression of the D antigen.
The Rh antigens are transmembrane polypeptides and are an integral
part of the RBC membrane. 3. Partial D (D Mosaic)
The gene products of RHD and RHCE are remarkably similar in that both One or more of the D epitopes within the entire D protein is either
encode for proteins composed of 417 amino acids that traverse the cell missing and/or is altered.
membrane 12 times and that their sequence differs by only 44 base Cells with a partial-D antigen usually type weaker than expected or may
pairs. not react at all when routine procedures are used with most commercial
anti-D reagents.

#MLSWATER2022 Page 4 of 9
MODULE 2 Part 3: Rh-Hr Blood Group System
DETERMINATION OF D STATUS or pregnancy. Rh antigens are highly immunogenic; the D antigen is the
Is required when testing donor bloods most potent.
Blood for transfusion is considered Rhpositive if either the D or the Exposure to less than 0.1 mL of Rh-positive RBCs can stimulate antibody
weak-D test is positive. production in an Rh-negative person.
Any donor blood sample that is typed Rh0(D)-negative by the slide or IgG1, IgG2, IgG3, and IgG4 subclasses of Rh antibodies have been
rapid tube method must be tested further by an indirect antihuman reported. IgG1 and IgG3 are of the greatest clinical significance because
globulin technique. the reticuloendothelial system rapidly clears RBCs coated with IgG1 and
IgG3 from the circulation.
NOTE: As with most blood group antigen sensitization, IgM Rh antibodies are
formed initially, followed by a transition to IgG.
If both tests are negative, the donor sample is considered Rh-
negative. Rh antibodies do not bind complement. For complement to be fixed (or
the complement cascade activated), two IgG immunoglobulins must
If the donor sample tests positive in any phase of Rh0(D) testing, the
attach to an RBC in close proximity. Rh antigens (to which the antibody
sample is considered Rh-positive.
would attach) are not situated on the RBC surface this closely.
Therefore, when an Rh antibody coats the RBCs, intravascular,
Determining the Rh0(D) status (including weak-D status) of obstetric
complement-mediated hemolysis does not occur.
patients is critical.
RBC destruction resulting from Rh antibodies is primarily extravascular.
All Rh-negative, weak D–negative obstetric patients are candidates for
Because Rh antibodies are primarily IgG and can traverse the placenta
Rh immune globulin (RhIg) (a drug injected to prevent Rh-negative
and because Rh antigens are well developed early in fetal life, Rh
individuals who are exposed to Rh-positive RBCs from developing
antibodies formed by Rh-negative pregnant women do cross the
antiD).
placenta and may coat fetal RBCs that carry the corresponding antigen.
o Likewise, when the mother is Rh-negative and the newborn is typed
Rh-negative, the weak-D status of the newborn must be determined This results in the fetal cells testing positive by the direct antiglobulin
to assess the likelihood of maternal sensitization and the need for test and in HDN, the coated fetal cells are removed prematurely from
Rh immune globulin prophylaxis for the mother. the fetal. Until the discovery of Rh immune globulin, anti-D was the
o If a newborn’s cells are coated with maternal IgG anti-D in utero, most frequent cause of HDN.
very few D antigen sites are available to react with reagent anti-D
(termed “blocking phenomena”). Rh Antigen Typing Reagents
Elution of the sensitizing antibody (removing the antibody) and The reagents used to type for D and for the other Rh antigens may be
identifying it as anti-D will verify that the infant’s RBCs are D-positive. derived from a variety of sources.
o The reagents may be high-protein–based or low-protein–based,
DETECTION OF RH ANTIBODIES AND ANTIGENS saline-based, chemically modified, monoclonal, or blends of
Rh Antibodies monoclonals.
Although the Rh system was first recognized by saline tests used to
detect IgM antibodies, most Rh antibodies are IgG immunoglobulins and Saline reactive reagents, which contain IgM immunoglobulin, were the
react optimally at 37 degrees C or after antiglobulin testing. first typing reagents available to test for the D antigen.
Rh antibodies are usually produced following exposure of the o Saline anti-D has the advantage of being lowprotein–based and can
individual’s immune system to foreign RBCs, through either transfusion be used to test cells that are coated with IgG antibody. The primary
disadvantages of saline typing reagents are their limited availability,

#MLSWATER2022 Page 5 of 9
MODULE 2 Part 3: Rh-Hr Blood Group System
cost of production, and lengthy incubation time. Because saline Hemolytic Disease of the Newborn (HDN)
anti-D is an IgM immunoglobulin, it cannot be used for weak-D HDN caused by Rh antibodies is often severe because the Rh antigens
typing. are well developed on fetal cells, and Rh antibodies are primarily IgG,
In the 1940s, high-protein anti-D reagents were developed. which readily cross the placenta.
o Human plasma containing high-titer D-specific antibody is used as After years of research, a method was developed to prevent susceptible
the raw material. (Rh0 D-negative) mothers from forming antiD, thus preventing Rh0(D)
o Potentiators of bovine albumin and macromolecular additives such HDN.
as dextran or polyvinylpyrrolidone are added to the source material
Rh-immune globulin, a purified preparation of IgG anti-D, is given to a D-
to optimize reactivity in the standard slide and rapid tube tests.
negative woman during pregnancy and following delivery of a D positive
o The presence of potentiators and the higher protein concentration,
fetus. Rh-immune globulin is effective only in preventing anti-D HDN.
however, increase the likelihood of false positive reactions.
To assess the validity of the high-protein Rh typing results, a control RH DEFICIENCY SYNDROME: RHNULL AND RHMOD
reagent was manufactured and had to be tested in parallel with each Rh
It is the rare individual who fails to express any Rh antigens on the RBC
test. If the control reacted, the test result was invalid and had to be
surface or exhibits a severely reduced expression of all Rh antigens. The
repeated using a different technique or reagent anti-D. The major
individuals who lack all Rh antigens on their RBCs are said to have the
advantages of high-protein anti-D reagents are reduced incubation time
Rhnull syndrome, which can be produced by two different genetic
and the ability to perform weak-D testing and slide typing with the same
mechanisms:
reagent.
o In the regulator-type Rhnull syndrome, there is a mutation in the
In the late 1970s, scientists chemically modified the IgG anti-D RHAG gene. This results in no Rh polypeptides or RHAG antigen
molecule by breaking the disulfide bonds that maintain the antibody’s expression on the RBCs, even though these individuals usually have
rigid shape. a normal complement of RHD and RHCE genes. These individuals
Monoclonal antibody reagents have become available more recently. can pass the normal RHD and RHCE genes to their children.
These reagents are derived from single clones of antibody-producing o In the second type of Rhnull syndrome (the amorphic type), there is
cells. a mutation in each of the RHCE genes and a deletion of the RHD
gene. The RHAG gene is normal.
CLINICAL CONSIDERATIONS Individuals with the Rhnull syndrome demonstrate a mild compensated
Transfusion Reactions haemolytic anemia, reticulocytosis, stomatocytosis, a slight-to-
Rh antigens are highly immunogenic. The D antigen is the most moderate decrease in haemoglobin and hematocrit levels, an increase
immunogenic antigen outside the ABO system. in hemoglobin F, a decrease in serum haptoglobin, and possibly an
Circulating antibody appears within 120 days of a primary exposure and elevated bilirubin level.
within 2 to 7 days after a secondary exposure. When transfusion of individuals with Rhnull syndrome is necessary, only
Rh-mediated hemolytic transfusion reactions, whether caused by Rhnull blood can be given.
primary sensitization or secondary immunization, usually result in
extravascular destruction of immunoglobulin-coated RBCs. NOTE:
The transfusion recipient may have an unexplained fever, a mild Individuals of the Rhmod phenotype have a partial suppression of Rh
bilirubin elevation, and a decrease in hemoglobin and haptoglobin. gene expression and exhibit features similar to those with the
The direct antihuman globulin test is usually positive, and the antibody Rhnull syndrome
screen may or may not demonstrate circulating antibody.

#MLSWATER2022 Page 6 of 9
MODULE 2 Part 3: Rh-Hr Blood Group System
However, the clinical symptoms are usually less severe and rarely A sample with the phenotype D C E c e can be either DcE/DCe or
clinically remarkable. Dce/DCE. Anti-rhi reacts only with Dce/DCe RBCs.
33 RBCs classified as Rhmod do not completely lack Rh or LW
antigens. G
Rhnull and Rhmod RBCs exhibit other blood group antigens; however, G is an antigen that is present on most D-positive and all Cpositive RBCs.
S, s, and U antigen expression may be depressed. In the test tube, anti-G reacts as though it were a combination of anti-C
34 Rhnull RBCs are negative for FY5. plus anti-D.
Was originally described in an rr person who received Dccee RBCs.
Subsequently, the recipient produced an antibody that appeared to be
anti-D plus anti-C, which should be impossible because the C antigen
UNUSUAL PHENOTYPES AND RARE ALLELES was not on the transfused RBCs.
Cw
Originally considered an allele at the C/c locus. Rh:13, Rh:14, Rh:15, Rh:16 Rh:13, Rh:14, Rh:15, and Rh:16
Later studies showed that it can be expressed in combination with both Define four different parts of the D mosaic, as it was originally
C and c and in the absence of either allele. described.
Now known that the relationship between C/c and Cw is only o Although these parts are included in the partial-D categories II to VII
phenotypic and that as defined by Tippett and Sanger, 25, 27 they are not directly
Antithetical to the high-incidence antigen MAR. comparable.
Found in about 2 percent of whites and is very rare in blacks.
Anti-Cw has been identified in individuals without known exposure to Hr0 Hr0
foreign RBCs as well as after transfusion or pregnancy. antigen present on all RBCs with the “common” Rh phenotypes (e.g.,
o Anti-Cw may show dosage (i.e., reacting more strongly with cells R1R1 , R2R2 , rr).40 When RBCs phenotype as D--, the most potent
from individuals who are homozygous for Cw). Because of the low antibody they make is often one directed against Hr0.
incidence of Cw, Cw antigen–negative blood is readily available.
Rh:23, Rh:30, Rh:40
f (ce) all low-frequency antigens associated with a specific category of partial-
The f antigen is expressed on the RBC when both c and e are present on D. Rh:23 (also known as Wiel and Dw) is an antigenic marker for
the same haplotype (i.e., cis position); it has been called a compound category Va partialD.41 Rh:30 (also known as Goa or Dcor) is a marker
antigen. for category Iva partial-D.42 Rh:40 (also known as Tar or Targett) is a
Phenotypically, the following samples appear the same when tested marker for category VII.
with the five major Rh antisera: Dce/DCE and DcE/DCe. However, when
tested with anti-f, only the former reacts. Rh:33
Anti-f has been reported to cause HDN and transfusion reactions. The low-incidence antigen Rh:33 is associated with a rare variant of the
R0 (Dce) gene called R0 Har. 44 R0 Har gene codes for normal amounts
rhi (Ce) of c, reduced amounts of e, reduced f, reduced Hr0, and reduced
Similar to f, rhi is present when C and e are in the cis configuration, has amounts of D antigen.
been called a compound antigen, and is a single entity. The D reactions are frequently so weak that the cells are frequently
mistakenly typed as Rh negative.
#MLSWATER2022 Page 7 of 9
MODULE 2 Part 3: Rh-Hr Blood Group System
To denote the weakened expression of an antigen in Fisher-Race o DC–, Dc–, D–E, D--. The antibody made by D-- people is called anti–
nomenclature the letter is placed in parentheses. The R0 Har gene Rh 17 or anti-Hr0.
expresses (D)c(e) and has been found in whites. o A variation has been recognized within the deletion D--, called D••.
Rh:32 The D antigen in the D•• is stronger than that in DC–, D–E, Dc–, or D–e
Rh:32 is a low-frequency antigen associated with a variant of the R1 samples but weaker than that of D-- samples.
[D(C)(e)] gene that is called RN. A low-incidence antigen called Evans (Rh:37) accompanies the Rh
The C antigen and e antigen are expressed weakly. structure of D•• cells.
The D antigen expression is exaggerated or exalted. This gene has been Transfusion of individuals with a Rh deletion or D•• phenotype is
found primarily in blacks. difficult if multiple antibodies are present; blood of a similar phenotype
would be required.
e Variants THE LW ANTIGEN
It appears, especially in the black population, that the e antigen may LW systems.
exhibit the same mosaic quality described for D. Because of these o Anti-LW reacts strongly with most D-positive RBCs, weakly
variations, e typings can be unreliable. (sometimes not at all) with Rh-negative RBCs, and never with Rhnull
Among the variants at the e locus are hrs, hrB, and VS(es ), with a cells.
variant R0 or r gene making e plus one or the other of these pieces. The independent segregation of LW from the Rh blood group genes was
Such variants are usually recognized when they make antibodies that established by a family study on a D-positive LW-negative woman; other
behave as anti-e, even though their RBCs type as e-positive with routine family studies support this point.
Rh typing reagents. There are three alleles at the LW locus: LWa , LWb , and LW (a silent
allele). Persons lacking LW antigen altogether are LW/LW and express
V, VS no LW on the RBCs.
The V(ceS) antigen is found in about 30 percent of randomly selected LWa is very common, and LWb is rare. When the Rhnull is present due
American blacks. to the suppression mechanism, the Rh and LW genes are not expressed
In selected individuals, it appears to be the serologic counterpart of f on the RBC; however, the Rh and LW genes are normal and, when
because it is present when c is cis with eS. passed to offspring, can be expressed normally.
(The VS(eS) antigen is also relatively common in blacks with the VS The amorphic Rhnull individual inherits the rr genes, which do not
antibody, reacting with all V-positive RBCs and additionally with r′s express Rh antigens; therefore, LW genes cannot be expressed.
RBCs.48) Although the relationship of V to VS remains somewhat less Anti-LW usually reacts more strongly with D-positive RBCs than with D-
than clear, both are markers associated with the black population. negative adult RBCs.
A weak anti-LW may react only with D-positive RBCs, and enhancement
Deletions techniques may be required to demonstrate its reactivity with
Dnegative cells.
There are very uncommon phenotypes that demonstrate no Cc and/or
Ee reactivity. Anti-LW reacts equally well with cord cells regardless of their D type.
Many examples lacking all Cc or Ee often have an unusually strong D This is an important characteristic to remember when trying to
antigen expression, frequently called exalted D. differentiate anti-LW from anti-D.
The deletion phenotype is indicated by the use of a dash (–), as in the Also, anti-LW more frequently appears as an autoantibody, which does
following examples: not present clinical problems.

#MLSWATER2022 Page 8 of 9
MODULE 2 Part 3: Rh-Hr Blood Group System
APPENDICES CLASS ACTIVITY
1. Patient is D+C+E+c and e was not tested
Wiener Fisher-Race Wiener Fisher-Race o Convert to Rosenfield: Rh: 1, 2, 3, -4
Ro Dce r dce How about when D is positive, C is negative, E is positive, c is positive
R1 DCe r’ dCe and e is negative?
R2 DcE r” dcE o Convert to Rosenfield: Rh: 1, -2, 3, 4, -5
Rz DCE R^y dCE
2. Convert to Fisher Race
ISBT FISHER-RACE WIENER ROSENFIELD o R1r= DCe/dce
004001 D Rho Rh1 o R1R1= DCe/DCe
o r1r1= dCe/dcE
004002 C rh’ Rh2
o R2ry= DcE/dCe
004003 E rh” Rh3
004004 c hr’ Rh4
3. Rh: 1, -2, 3, 4 ,5
004005 e hr” Rh5
o What is the Phenotype? D+C+E+c+e or DcE/ce
o Convert to Weiner: R2r
COMMON Rh TYPES BY THREE NOMENCLATURES
COMMON GENOTYPE 4. Wiener to Rosenthal
WIENER FISHER-RACE ROSENFIELD FREQUENCY (%) o R1r= Rh: 1, 2, -3, 4, 5
(approx., White) o R2R0= Rh: 1, -2, 3, 4, 5
R1r DCe/dce Rh: 1,2, -3, 4, 5 33 o RzR1= Rh: 1, 2, 3, -4, 5
R1R1 DCe/DCe Rh: 1, 2, -3, -4, 5, 18 o ryr= Rh: -1, 2, 3, 4, 5
rr dce/dce Rh: -1, -2, -3, 4, 5, 15
R1R2 DCe/DcE Rh: 1, 2, 3, 4, 5, 11 5. An Individual’s red blood cell give the following reactions with
R2r DcE/dce Rh: 1, -2, 3, 4, 5, 9 antisera:
R2R2 DcE/DcE Rh: 1, -2, 3, 4, -5, 2
RARER GENOTYPES Anti-D 4+
r’r dCe/dce Rh: -1, 2, -3, 4, 5, 1 Anti-C 3+
r’r’ dCe/dCe Rh: -1, 2, -3, -4, 5, 0.01 Anti-E 0
r”r dcE/dCe Rh: -1, -2, 3, 4, 5, 1 Anti-c 3+
r”r” dcE/dcE Rh: -1, -2, 3, 4, -5, 0.03 Anti-e 3+
R0r Dce/dce Rh: 1, -2, -3, 4, 5, 2 Rh control 0
R0R0 Dce/Dce Rh: 1, -2, -3, 4, 5, 0.1
ryr dCE/dce Rh: -1, 2, 3, 4, 5, rare The Individual’s most probable genotype is: Dce/dce
Rosenthal: 1, 2, -3, 4, 5

-END OF TRANSCRIPTION -
Kung na kaya mo si Complement System, makaya mo man si Rh Typing.
Kung indi, basta kaya mo na ah.

#MLSWATER2022 Page 9 of 9
MODULE 3: MINOR BLOOD SYSTEMS
MLS 18, March 13, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`

Legend: LEWIS SYSTEM


Transcription Bullet The gene for the Lewis system is linked to the third component of
Notes/Module Packet Bullet complement and is located on chromosome 19.
❖ PPT
The Lewis system differs from most other blood group systems in that
MODULE OUTLINE the Lewis genes must interact with Hh and secretor genes in order for
I. Systems that produce Cold-reacting Antibodies Lewis antigens to be produced
A. Lewis System
B. I Blood Group Collection ❖ Among all the blood group system, Lewis blood group is not
C. P blood Group manufactured by the RBC.
D. MNSs Blood Group System ❖ It is manufactured by tissue cells
II. Systems that Produce Warm-reacting Antibodies ❖ Type 1 glycosphingolipid that are possibly absorbed into the RBC
A. Kell System membrane
B. Kidd Blood System
C. Duffy blood System LEWIS ANTIGENS
III. Other Blood Group Antigens Lewis antigens are probably synthesized in the intestinal epithelium,
A. Lutheran Blood Group System are soluble in body fluids, and the antigens present in plasma are
IV. Clinical Significance of Antibodies of Minor Blood Groups adsorbed onto red cells.
LEARNING OUTCOMES The Lewis antigenic specificity is determined by the carbohydrate
1. Describe the characteristics of the antigens of the different minor fucose.
blood groups systems. The exact Lewis antigen produced is determined by the presence or
2. Describe the characteristics antibodies of the different minor blood absence of the secretor gene (Se).
groups systems and their clinical significance. Lewis antigens are easily and conveniently detected in human saliva by
3. Perform laboratory tests for the detection and identification of hemagglutination inhibition.
antigens &/or antibodies of the minor blood group systems Lewis antigens have also been detected in, and isolated from, human
milk, gastrointestinal juices, urine, seminal fluid, ovarian cyst fluid, and
amniotic fluid.
Leᵃ and Leᵇ cannot usually be detected on cord red cell samples by
SYSTEMS THAT PRODUCE COLD -REACTING ANTIBODIES direct agglutination.
A total of 30 blood group systems are recognized by the International Lewis antigens start to appear on red cells soon after birth.
Society of Blood Transfusion (ISBT) Lewis antigens in the saliva of neonates with an Le gene are the same
Nine blood group systems (ABO, Rhesus, Kell, Kidd, Duffy, MNS, P, as those detected in adult salivas
Lewis, and Lutheran) are considered to be clinically significant as Agglutinability of red cells with anti-Leᵃ or anti-Leᵇ may be reduced
these are known to cause hemolytic transfusion reactions (HTR) and during pregnancy and pregnant women with a transient Le(a–b–)
hemolytic disease of fetus and newborn (HDFN). phenotype may produce Lewis antibodies
#MLSWATER2022 Page 1 of 18
MODULE 3: MINOR BLOOD SYSTEMS
LEWIS ANTIBODIES
❖ Lewis antigenic specificity Lewis antibodies are common, usually react below body temperature,
o Determined by the carbohydrate fucose. are mainly or entirely IgM, and are not clinically significant
o Lewis gene will code for fucosyltranferase enzyme Lewis antibodies that are only active in vitro at temperatures below
• Fucosyltranferase enzyme can catalyse the addition of 37°C do not cause increased clearance of antigen-positive transfused
fucose to the 4th carbon of N-acatylgalactosamine of the red cells in vivo
type 1 precursor substance Lewis antibodies do not cause serious hemolytic disease of the
newborn, presumably because Lewis antigens are present in fetal
❖ Antigen in Lewis Blood Group System secretions, but generally not on fetal red cells.
o Do not develop as an integral part of the RBC membrane

❖ Expression in Lewis Blood Group System 1. Anti-Leᵃ


o The amount of antigen present is variable in partially dependent on 2 Types:
individuals ABO, phenotype and pH.
• Because Lewis antigen expression is affected by H gene, Human anti-Leᵃ
secretor gene and Lewis secretor gene o Leᵃ antibodies are usually ‘naturally occurring’ and predominantly
o May decrease dramatically during pregnancy, most especially after IgM
delivery o Agglutination of red cells with anti-Leᵃ is generally strongest at low
temperatures, often not occurring at all at 37°C.
4 Common Phenotype o Anti-Leᵃ usually fix complement
Lewis A+B- Lewis A, presence of A positive o A two-stage complement addition antiglobulin test is often a
Are mostly non secretors of the ABH successful way of detecting anti-Leᵃ
Lewis A- B- Lewis B, presence of B positive
Secretors of ABH It can best react in room temperature but can also react in 37°C
Although most of Lewis antibodies are considered to be clinically
Lewis A+ B+ Lewis AB, presence of A,B positive
insignificant but they can have the ability to bind with the
Frequently seen in Africans
complement therefore it can trigger in-vitro hemolysis
Lewis A- B- Lewis null, it does not contain any antigen
Anti-Leᵃ are not associated with hemolytic disease of the newborn
Frequently seen in Asians
since there is no presence of anti Le a in the fetal RBC
The antibodies that would react with 37°C, that can trigger the
❖ Most neonates/newborns/infant are Lewis A-B- regardless of which
complement and capable also in triggering in-vitro hemolysis can be
Lewis gene is inherited
associated with the hemolytic transfusion reaction
❖ When the neonates/newborns/infant contain Lewis gene and secretor
anti-Le a can trigger hemolytic transfusion reaction by triggering in-
gene, the A- will become A+ in 2 weeks – 6 months
vitro hemolysis
❖ A+ with the presence of secretor gene can change to B+
Anti-Leᵃ can be enhanced by enzyme treatment
❖ Non secretor will have A+B- but if secretor gene is present A-B+ is the
Lewis gene
Monoclonal anti-Leᵃ

#MLSWATER2022 Page 2 of 18
MODULE 3: MINOR BLOOD SYSTEMS
2. Anti-Leᵇ Ii ANTIGENS
Human anti-Leᵇ Clausen and Hakomori refer to I and i as histoblood group antigens
Like anti-Leᵃ, anti-Leᵇ are found most often in people with Le (a– b–) because, like ABH, Ii antigens are not restricted to red blood cells, but
red cells; unlike anti-Leᵃ, these people are generally non secretors of are usually found on the surface of most human cells and on soluble
ABH glycoproteins in various body fluids.
Anti-Leᵇ are generally IgM and would react best in room temp. I and i antigens have been detected in amniotic fluid, urine, and
❖ They can bind oppositely with Anti-Leᵃ ovarian cyst fluid
I and i are present on lymphocytes from cord and adult blood. Anti-I
It binds with complement poorly and -i are potent cold lymphocytotoxins, effective at killing B and T
Activities are enhance by enzyme treatment lymphocytes
Not associated with hemolytic disease of the newborn The i antigen is a characteristic of dividing cells present on a variety of
can cause hemolytic transfusion reaction very rarely cell types including lymphoblasts, fibroblasts, erythroblasts, and
may appear transiently during pregnancy in Lewis A-B- women, but thymocytes
disappears after delivery Ii antigens represent developmental antigens on red cells and in many
other tissues
Animal anti-Leᵇ
Polyclonal and monoclonal Anti-ALeᵇ 1. I antigen (I1)
Anti-A1Leᵇ, an antibody that reacts only with the red cells of A1 Lewis- I antigen can be detected in saliva by hemagglutination inhibition
positive secretors [A1 Le(a–b+)], was first found in the serum of a
I is present in human milk in greater quantities than in saliva and
group A1B Le(a–b–) man
most anti-I are inhibited to some extent by milk
I antigen is subdivided into:
I BLOOD GROUP COLLECTION
Cold agglutination involves the occurrence and reaction of o Iᴰ (developed)
autoantibodies, reacting optimally in the cold (0°C) with red blood • Is the I antigen on most adult cells, but not cord or adult i
cells. These autoantibodies are termed cold agglutinins. cells;
Low titer cold agglutinins are present in the sera of all adults. The most “D” for developed; usually seen in adults
prevalent of these autoantibodies is a heterogeneous assembly of poorly developed at birth, I antigenic strength increases = i
specificities called anti-I. antigenic strength decreases
Anti-I are generally weak, but potent examples may be found as
autoantibodies in patients with cold haemagglutinin disease (CHAD) or o Iᶠ (fetal)
following Mycoplasma pneumoniae infection • Is the I antigen expressed on all human red cells including
The i antigen has a reciprocal relationship with I cord and adult i cells. Iᴰ therefore has a reciprocal
I blood group antigens (I and i) are structurally-related with the ABO relationship with i.
antigens; found in the RBC membrane, in the plasma in the milk, and • “f” for fetal; usually seen with newborns
even in the amniotic fluid • Antigenic strength would only be seen with neonates or
infants
Treatment of red cells with proteases or with sialidase generally
enhances expression of Ii antigens.

#MLSWATER2022 Page 3 of 18
MODULE 3: MINOR BLOOD SYSTEMS
2. i Antigen (I2) associated with infectious mononucleosis, may also be associated with
hemolytic anemia (disappears as the infection is resolved), can be
3. Iᵀ antigen associated w HDN, hemolytic anemia, & infectious mononucleosis
Iᵀ is the name given by Booth et al. to an antigen detected by cold
agglutinins present in the sera of a high proportion of Melanesians 3. Anti-Iᵀ
and expressed strongly on cord cells, weakly on normal adult cells, Anti-Iᵀ defines an antigen expressed strongly on cord cells, weakly on
and weaker still on adult i cells. most adult cells, and very weakly on adult i cells
The ‘T’ stands for ‘transition’ as Iᵀ was assumed to represent a
developing I antigen at the transition from I to i 4. Anti-j
These antibodies reacted with protease- and sialidase-treated red
Ii ANTIBODIES cells, but not with cells treated with endo-b-galactosidase
They resembled most pathogenic anti-I and -i by expressing the 9G4
1. Anti-I idiotype, a characteristic of antibodies encoded by a gene utilizing a
3 Types: V4- 34 sequence

Autoanti-I
o These antibodies directly agglutinate I-positive red cells at 4°C with
P BLOOD GROUP
varying thermal amplitude, but are generally inactive above 30°C
Naturally-occurring, “reacting IgM antibodies”, fail to react w cord P antigenic determinants on red cells reside in the carbohydrate
RBCs, some react in a broader temp range, and can cause cold residues of glycosphingolipids, oligonucleotide chains attached to
agglutinin disease, cannot bind or be associated with HDN; ceramide
related/associated w mycoplasma pneumoniae which causes P antigens are structurally-related to ABO antigens; exist as glycolipid
mycoplasma pneumonia (primary atypical pneumonia) or glycoprotein

Common Phenotypes:
Alloanti-I
o It does not react with autologous cells.
P1 • One of the most (out of two) common phenotypes
o These antibodies are almost invariably IgM and usually only active involved; approx. 75% of the population have this
at low temperatures. o P1 individuals have both P and P1 antigens
o P antigen is well developed at birth while P1
Anti-I lectin antigen is poorly developed
o Lectin prepared from the gonads of Aplysia depilans, a marine • The P expression in adult is wildly variable
mollusc (sea slug), behaved serologically as anti-I P2 • The other most common phenotype, individuals with
this phenotype have only P antigen in their RBC.
2. Anti-i P1k • Is very rare, individuals have both P1 and PK antigen
in their rbc
Anti-i are heterogeneous in specificity
Anti-i is often found in the serum of patients with infectious P2k- • Also rare, have both Pk and P2 antigens in their rbc
mononucleosis and occasionally causes hemolysis P • Extremely rare, rbc with this phenotype, negative for
The presence of anti-i is associated with immunodeficiency P, P1, and Pk antigen.

#MLSWATER2022 Page 4 of 18
MODULE 3: MINOR BLOOD SYSTEMS
P ANTIGENS Monoclonal anti-P1
1. P1 antigen
P1 on red cells is a glycosphingolipid 2. Anti-P
Anti-P is found in the serum of all Pᴷ individuals and can be separated
2. Pᴷ antigen from serum of p individuals by adsorption with P1ᴷ or P2ᴷ cells, or by
Pᴷ was present on the fibroblasts of all P1 and P2 individuals, and only inhibition with HCF
absent from those of p people 3 Types:
Pk (also known as CD77) has been detected on lymphocytes,
granulocytes,monocytes, platelets, smooth muscle of the digestive Alloanti-P
track and urogenital system, and in other tissues. Are produced by individuals with PK1 and PK2 phenotypes. Can
Pk is also expressed on malignant cells and cell lines derived from them trigger severe hemolytic transfusion reaction.
and is a useful marker for Burkitt’s lymphoma Also associated with Donath–Landsteiner (DL) antibodies causes
cold reacting antibody associating with paroxysmal cold
3. P antigen (globoside) hemoglobinuria
Globoside is the most abundant red cell membrane glycolipid
P antigen, the glycolipid globoside , is found on all red cells except Donath–Landsteiner (DL) antibodies
those of the rare phenotypes p and Pk o Generally, have P specificity
P antigen is found on malignant cells and cell lines derived from them. o Paroxysmal cold hemoglobinuria (PCH) is a rare form of
P has also been detected on fetal liver, fetal heart, and on placenta autoimmune hemolytic anemia (AIHA), occurring predominantly in
young children following viral infections.
4. LKE antigen o Sera from patients with PCH usually give a positive Donath–
Landsteiner test; i.e. the antibody binds in the presence of
complement at 0°C and hemolyses the cells when subsequently
warmed
P antibodies
Anti-P1 Monoclonal anti-P
Are naturally occurring antibodies, are called reacting IGM, often
seen with individuals with P2 phenotype.
3. Anti-Pᴷ
They rarely react at high room temperature but bind with
3 Types:
compliment. Do not cause Htn but rarely associated with hemolytic
o Alloanti-Pᴷ
transfusion reaction. Can be neutralized using hyatid cyst fluid from
o Autoanti-Pᴷ
echinococcus granulosus infection.
o Monoclonal anti-Pᴷ
3 Types:
• A monoclonal antibody was shown to define the neutral
glycolipid
Alloanti-P1
o Alloanti-P1 is a common specificity, usually as a weak agglutinin
active only at low temperature.

Animal anti-P1
#MLSWATER2022 Page 5 of 18
MODULE 3: MINOR BLOOD SYSTEMS
MNSs BLOOD GROUP SYSTEM 1. Mᵍ
The second blood group system discovered, is probably second only to a very rare antigen first described by Allen et al. in 1958, is encoded by
Rh in its complexity a gene that produces virtually no M or N antigen.
M and N determinants are carried on glycophorin A (GPA), the major Like M and N, Mᵍ is denatured by treatment of the cells with trypsin,
red cell sialic acid-rich glycoprotein (sialoglycoprotein, SGP). but not chymotrypsin; unlike most anti-M and -N, anti-Mᵍ generally
M differs from N in the amino acid composition of the extracellular tip react with sialidase-treated Mᵍ + cells
of GPA: Anti-Mg is easily inherited by the glycosylated N terminal octapeptide
M has serine at position 1 and glycine at position 5; cleaved from GPAMg, but not by that from GPAN
N has leucine at position 1 and glutamic acid at position 5 Mᵍ is extremely rare, yet anti-Mᵍ is possibly the most common MNS
S and s are carried on another red cell SGP called glycophorin B (GPB). antibody
GPB, another sialic acid-rich red cell membrane glycoprotein, is closely
related in structure to GPA 2. Mᶜ
The S/s distinction arises from a single amino acid substitution at Cannot strictly be regarded as a blood group antigen as no anti-Mᶜ
position 29 of GPB: exists.
S has methionine at position 29 Mᶜ is often considered to represent an intermediate between M and N
s has threonine at position 29 Rare allele of M and N found by Dunsford et al. in an English family,
Anti-M & Anti-N antibodies produces a determinant that reacts with the majority of anti-M (as
IgM saline reaction demonstrated by the red cells of the N/Mᶜ member of the family) and
Dosage effect with the minority of rabbit anti-N (as demonstrated by the two M/Mᶜ
Possible HDN & HTR if reaction at AHG members).
subsequently been defined by a pattern of reactions with known anti-
M and -N reagents
M and N antigens
The amino acid sequence of GPA demonstrates polymorphic variation NOTE:
at positions 1 and 5, represented serologically as the MN blood groups. Trypsin catalyzes the hydrolysis of peptide bonds on the carboxyl
GPA isolated from M+N– individuals has serine at position 1 and side of lysine and arginine residues.
glycine at position 5 Chymotrypsin (a-chymotrypsin) normally hydrolyses the peptide
GPA from M–N+ individuals has leucine at position 1 and glutamic acid bond on the carboxyl side of the aromatic amino acids
at position 5. phenylalanine, tryptophan, and tyrosine, as well as leucine,
Both forms of GPA can be isolated from M+N+ individuals. methionine, asparagine, and glutamine
The terminal serine of M-active GPA is not glycosylated; amino acid Papain, ficin, and bromelin are enzymes, and have a rather broad
residues 2, 3, and 4 of GPAM and GPAN are O-glycosylated. specificity and the preparations available are often crude compared
Various proteolytic enzymes have proved very useful in the serological with trypsin and chymotrypsin. Most GPA- and GPB borne antigens
identification, analysis, and definition of antigens belonging to the MNS are destroyed by treatment of red cells with these enzymes, only
system those situated close to the red cell membrane survive.
GPA and GPB carry about 15 and 11 O-linked oligosaccharides,
respectively, most of which contain two molecules of sialic acid. In
addition, GPA has one N-glycan, which may also be sialylated.

#MLSWATER2022 Page 6 of 18
MODULE 3: MINOR BLOOD SYSTEMS
Sialidase (neu-raminidase) treatment of red cells removes at least U–red cells are almost always S–s–, but S–s– cells are often U+.
some of these sialic acid residues, altering the charge and possibly S–s–U+ is often referred to as S–s–U+var.
the shape of the molecules. Strength of U antigen expression on S–s–U+ red cells is variable;
adsorption–elution tests or sensitive agglutination tests with a
particularly potent antibody may be required for its detection
Most monoclonal anti-M and -N do not react, or react comparatively anti-U is unclear, but the term is traditionally used to describe
weakly, with desialylated red cells or isolated glycophorins antibodies produced by S–s– individuals to high-frequency
determinants on GPB
Unusual MNS phenotypes caused by two very rare gene deletions: S–s–U– and S–s–U+var cells usually lack the trypsin-resistant N antigen
carried on GPB (‘N’)
U antigen is generally resistant to denaturation by sialidase, trypsin,
En(-a)
chymotrypsin, papain, and ficin. However, unusual examples of anti-U
deletion of the coding region of GYPA, causes a deficiency of GPA,
do not react with papain-treated cells
but not GPB.
S–s–U– red cells do not show most of the unusual serological reactions
En(a–) cells have normal or enhanced expression of S and/or s
associated with reduced sialic acid that are characteristic of red cells
Anti-Ena represents an umbrella term, which describes antibodies
deficient in GPA , though Glycine soja lectin may agglutinate U-
to determinants on various parts of GPA.
deficient cells
The En(a–) phenotype can arise in a number of ways. Typically,
S–s–U– red cells are deficient in GPB. This has been demonstrated by
En(a–) represents homozygosity for a rare gene deletion (called En
failure to inhibit anti-S, -s, or -U with sialoglycoproteins isolated from
for convenience) at the GYPA locus, resulting in no production of
S– s–U– cells, by SDS PAGE of red cell membranes or isolated
GPA, but normal production of GPB.
sialoglycoproteins , and by immunoblotting with antibodies and lectins
directed at determinants on GPB
Mᴷ
a deletion of the coding regions of GYPA and GYPB is responsible
for deficiency of GPA and GPB MNSsU ANTIBODIES
The name Mᴷ was coined by Metaxas and Metaxas-Bühler for a 1. Human anti-M
new allele of M and N that appeared to produce neither M nor N Most anti-M are only reactive at temperatures below 37°C, with an
Mᴷ produces neither GPA nor GPB; red cells from Mᴷ/Mᴷ optimum temperature of 4°C, but occasional examples will agglutinate
homozygotes are devoid of GPA and GPB . Red cells of people red cells at body temperature
heterozygous for Mᴷhave about half the normal quantity of GPA Most human anti-M contain an IgM component, although 78% were
and GPB found to be at least partially IgG and these IgG antibodies could
agglutinate saline suspensions of M+ red cells.
S AND S ANTIGENS Naturally occurring, may be both IgM and IgG, do not bind w a
The S/s polymorphism is represented by a single amino acid complement, react at room temp or lower, rarely associated w HDR &
substitution in GPB at position 29; S-active GPB has Met29 and the s- HTR, some are pH-dependent (best at 6.5) and glucose-dependent
active molecule has Thr29
S/s genotyping on genomic DNA can be carried out by PCR-based 2. Human anti-N
techniques with an allele-specific primer Red cells of individuals with the rare M+N–S–s–(U– or U+var)
S, s, and most other MNS system antibodies are not sialic acid- phenotypes lack ‘N’ and may produce a potent alloanti-N, which will
dependent
#MLSWATER2022 Page 7 of 18
MODULE 3: MINOR BLOOD SYSTEMS
agglutinate all cells carrying an N determinant, whether on GPA or SYSTEMS THAT PRODUCE WARM-REACTING ANTIBODIES
GPB. KELL SYSTEM
These antibodies have been referred to as anti‘N’, -N‘N’, or -NU
Most anti-N are ‘naturally occurring’, IgM, and inactive above 25°C Kell antigen
None of the Kell antigens is expressed on cells of the Kell-null
Rare, weak, naturally-occurring IgM antibodies, react at room temp or
phenotype, Ko, which arises from homozygosity for a silent gene at the
below, usually not associated w HDN & HTR KEL locus.
MNSs antibodies follow the dosage effect; homozygous = stronger Ku antigen is present on all cells save those of the Ko phenotype
agglutination compared to heterozygous The Kell antigens are located on CD238, a red cell transmembrane
glycoprotein of apparent Mr 93000, a metalloendopeptidase
3. Anti-S The molecular bases for most of the Kell antigens are known and all the
S, s, and U antibodies usually react at 37°C, but most are optimally Kell system polymorphisms result from single amino acid substitutions.
reactive at temperatures between 10 and 22°C by manual antiglobulin KEL is situated on chromosome 7q32–q36
tests under normal ionic conditions McLeod syndrome is a form of neuroancanthocytosis, which includes
Anti-S reagents are notorious for containing antibodies to private an abnormal Kell red cell phenotype.
antigens. McLeod phenotype red cells have depressed Kell antigens and lack the
Anti-S has been implicated in haemolytic transfusion reactions and has high frequency antigen Kx.
caused severe and fatal HDN The inheritance of Kx is controlled by an X-borne gene, XK, and
Autoanti-S has been responsible for AIHA represents a blood group system (the Kx system) independent of Kell.
Autoantibodies that are probably detecting nonpolymorphic The Kx protein and Kell glycoprotein are linked by a disulphide bond
determinants on GPB may ‘mimic’ anti- S because of the greater Kell antigens are well developed at birth. K was found in fetuses of 10–
quantity of GPB molecules on S+ cells than on S– cells 11 weeks’ and k at 6–7 weeks’ gestation
K18 is the only Kell antigen not shown to be inherited.
4. Anti-s TOU (KEL26) is an antigen of high frequency absent from Ko cells and
Anti-s are usually optimally reactive at 22°C or below shown to be located on the Kell glycoprotein by a MAIEA analysis
Anti-s has been responsible for severe and fatal HDN and for a delayed Kell system antigens are destroyed by disulphide-bond reducing agents
haemolytic transfusion reaction and so must be dependent on the native conformation of the molecule
Compromised of 21 high and low incidence antigen. The most
significant of which are the K and k antigen.
5. Anti-U
KO phenotype - lack all K antigen and the antigens associated with Kell
Anti-U are generally non-complement-binding IgG antibodies
system.
containing an IgG1 component
Kx antigen – located in the x chromosome, rbc that lacks Kx antigen
no ‘naturally occurring’ anti-U has been reported.
also have greatly weaken expression of other kell antigen. Rbc are
Like anti-S and -s, U antibodies may have greater reactivity at
morphologically acanthocytic they have decrease survival and less
temperatures below 22°C than at body temperature
permeable to water and this syncdrome is called Mcleod syndrome.
“U” = universal; usually reactive in the anti-globulin phase, rare and
Mcleod Syndrome – characterized by splenomegaly with
occurs in S-s-U-people, Black origin (African black)
reticulocytosis and occasional associated with chronic granulomatous
disease.
Kell antigens are inactivated by sulfhydryl reagents.

#MLSWATER2022 Page 8 of 18
MODULE 3: MINOR BLOOD SYSTEMS
The Kx system 5. Anti-Kpᵇ
The Kx system consists of one antigen, Kx (XK1 or 019001), The first anti-Kpᵇ(Rautenberg) was found during routine
encoded by an X-linked gene, XK. crossmatching ; the serum also contained antiK
Absence of Kx from red cells results in severe reduction in Anti-Kpᵇ has been responsible for a delayed haemolytic transfusion
expression of Kell antigens, the McLeod phenotype. reaction

KELL ANTIBODIES 6. Anti-Kpc


The first anti-Kpc , called anti-Levay for 34 years, was made by a
1. Anti-K
patient with lupus erythematosus diffusus in response to transfusion
Anti-K is a relatively common cause of severe hemolytic disease of the
newborn (HDN)
Anti-K can be responsible for severe hemolytic transfusion reactions, 7. Jsa and Jsb (KEL6 and KEL7)
including reactions caused by incompatibility between donations given The Jsa /Jsb polymorphism is associated with two nucleotide changes
to the same patient in exon 17 of KEL, one encoding an amino acid substitution: Jsa , C1910
Pro597, G2019 Leu633; Jsb, T1910 Leu597, A2019 Leu633
2. Alloanti-K
Anti-K is the most common immune red cell antibody outside of the 8. Anti-Jsᵃ
ABO and Rh systems Anti-Jsa generally react best by the antiglobulin test and are red cell
Anti-K is often found in sera containing antibodies to high incidence immune in origin
Kell system antigens.
Anti-K, like other Kell system antibodies, are generally IgG, and 9. Anti-Jsᵇ
predominantly IgG1 All examples of anti-Jsᵇhave been found in black people.
They generally work best by the antiglobulin test.
3. Anti-k Anti-Jsᵇhas caused severe HDN , resulting in fatal hydrops fetalis
Most anti-k are IgG (often IgG1 ) and work best by the antiglobulin test,
but cold agglutinating IgM anti-k are known 10. Anti-Ku (-KEL5)
Anti-k has been responsible for haemolytic transfusion reactions, Anti-Ku is the typical antibody of immunized Ko individuals and detects
including severe intravascular haemolysis, and for HDN an antigen present on all red cells apart from those of the Ko
Considered to only be second to D antigen in autoimmunity. Usually phenotype.
react best in 37 degree Celsius. May occasionally bind with the It appears to be a single specificity and cannot be separated, by
compliment and may cause hemolytic disease to newborn and adsorption and elution, into components of other Kell specificity
haemolytic transfusion reaction The original anti-Ku caused both HDN and a haemolytic transfusion
reaction.
4. Anti-Kpᵃ Anti-Ku was also responsible for a severe transfusion reaction resulting
Many examples of anti-Kpa are known. The first (Penney) appeared to in jaundice, renal failure, and anuria
be ‘naturally occurring’ but, as with most anti-Kpa, reacted best by the
antiglobulin test.
Anti-Kpa very rarely causes HDN severe enough to require transfusion

#MLSWATER2022 Page 9 of 18
MODULE 3: MINOR BLOOD SYSTEMS
KIDD BLOOD GROUP SYSTEM Anti - Jkᵃ has been responsible for severe and fatal immediate HTRs
Jkᵃ and Jkᵇ of the Kidd system are the products of alleles and are and is regularly associated with delayed HTRs, which may be severe,
polymorphic in all populations tested. leading to oliguria, renal failure, and even death.
Both jka and jkb may exhibit dosage effect. Reactivity may also be Anti - Jkᵇ has also been incriminated in severe delayed HTRs
enhanced with enzyme treatment. Poorly immunogenic and can cause A major reason why Kidd antibodies are such a common cause of
severe haemolytic transfusion reaction. Both are noted for causing delayed HTRs is their tendency to fall rapidly to low or undetectable
delayed haemolytic transfusion reaction. Would react best in 37 levels in the plasma
degrees Celsius.
Most of Kidd Antibodies are Igg1 and Igg3 that’s why they can bind 2. Anti-Jkᵃ and -Jkᵇ Autoantibodies
complement efficiently and they would cross the placenta and cause
mild to severe hemolytic transfusion reaction. Very hard to detect, 3. Anti-Jkᵃ and -Jkᵇ Monoclonal antibodies
enzymes are used (LISS, PEG) IgM human monoclonal anti - Jkᵃ and - Jkᵇ have been produced by
Kidd antibodies are often difficult to work with. They are potentially Epstein - Barr virus - transformation of lymphocytes from immunised
dangerous, as they are a common cause of delayed haemolytic donors and fusion with mouse myeloma cells to form
transfusion reactions. heterohybridomas
A rare null phenotype, Jk(a–b–), is generally inherited recessively and
is most commonly found in Polynesians.
4. Anti - Jk3 (Jk3 Antibodies)
o Jk(a–b–) cells lack the high incidence antigen Jk3.
react optimally by an antiglobulin test, the reaction being enhanced by
o Jk(a – b – ) red cells lack the Kidd glycoprotein
enzyme treatment of the cells.
The Kidd glycoprotein functions as a urea transporter
Enzyme -treated cells may be hemolyzed by anti - Jk3 in the presence
The JK (SLC14A1) locus is on chromosome 18 at 18q11–q12
of fresh serum
Jkᵃand Jkᵇ are inherited as codominant alleles.
Anti - Jk3 are usually IgG
Jka, Jkb, and Jk3 are papain-, ficin-, trypsin-, chymotrypsin-, and
Anti - Jk3 may decline rapidly in vivo
pronase-resistant
Anti - Jk3 has been responsible for severe immediate and delayed
HTRs.
Kidd antibodies Most babies of mothers with anti - Jk3 are clinically unaffected,
1. Anti-Jkᵃ and -Jkᵇ Alloantibodies although the baby’s red cells may give a positive DAT, and in a few
Generally, an antiglobulin test is required to detect Kidd antibodies. cases phototherapy was administered
Kidd antibodies are usually IgG or a mixture of IgG and IgM; they are
rarely pure IgM
Anti - Jkᵃ detectable only by the manual Polybrene test was responsible
for an HTR, emphasizing the importance of detecting weak Kidd
antibodies.
Many anti - Jkᵃ react more strongly with Jk(a + b – ) than with Jk(a + b
+ ) cells, and some anti - Jkᵃ can only be detected with Jk(a + b – ) cells
Kidd antibodies, which are often difficult to detect, are a hazard in
blood transfusion.

#MLSWATER2022 Page 10 of 18
MODULE 3: MINOR BLOOD SYSTEMS
The Kidd glycoprotein is the red cell urea transporter, UT -B Fyᵃ and Fyᵇ are fully developed at birth and have been detected on red
The Kidd glycoprotein is present on endothelial cells of the vasa cells from embryos as early as 6 – 7 weeks gestation
recta, the vascular supply of the renal medulla, but is not present in The expression of Fyᵃ and Fyᵇ is as strong on red cells of very young
renal tubules. fetuses as on those of adults, and remains unmodified throughout fetal
Urea transporters in the kidney play an important role in life
concentrating urea in the renal medulla, whilst conserving water, in In addition to its presence on red cells, DARC, detected with anti-Fy6, is
order to produce concentrated urine abundant on endothelial cells lining post - capillary venules throughout
UT - B in human colon epithelium could participate in the transport the body, except for the liver , and on Purkinje neurons of the
of urea across the colon mucosa and assist in the maintenance of a cerebellum
normal colonic bacterial population Fyᵃ and Fyᵇ are not present on lymphocytes, monocytes, granulocytes,
2 Main Functions: or platelets
transporting urea rapidly in and out of the cells to prevent Use of impure preparations of trypsin containing chymotrypsin
shrinkage as they pass through the high urea concentration of the probably accounts for some early reports that Duffy antigens are
renal medulla, and to prevent swelling as they leave; trypsin - sensitive.
to prevent the red cells from carrying urea away from the renal Sialidase treatment of red cells does not affect the activity of Duffy
medulla, which would decrease the urea concentrating efficacy of antigens.
the kidney
-Please refer to reference book for the missing information (On Jk antigens & 2. FYx
antibodies) The presence of Fyˣ may be confirmed serologically by adsorption and
elution of anti - Fyᵇ
Fyˣ behaves as a weak Fyᵇ antigen; there is no anti – Fyˣ.
DUFFY BLOOD GROUP
Has 4 alleles that are important for the major antigen and are resulting 3. Fy3
from the type. Fy3 is present on all red cells apart from those of the Fy(a – b – )
Duffy A and B are produced by codominant alleles phenotype.
FYx is a weaken form of Duffy b Fy3 is a public antigen in people of European and Asian origin,
FY are produces no gene factor polymorphic in many black populations, and a private antigen in some
Fy (A-B-) would have resistance from the infection of Plasmodium parts of Africa.
vivax. In contrast to Fyᵃ and Fyᵇ, Fy3 is resistant to the treatment of red cells
with proteases
Duffy Antigens Red cells of some primates have Fy3, but not Fyᵃ or Fyᵇ
1. Fyᵃ and Fyᵇ
Fy a and Fyb result from a Gly42Asp substitution within the Duffy 4. Fy5
glycoprotein. Fy5 closely resembles Fy3
Fyᵃ and Fyᵇ are very sensitive to most proteolytic enzymes: it differs by its absence from Fy3 - positive Rh null cells (amorph and
They are completely destroyed by treatment of the red cells with regulator type), weak expression on red cells of D – – phenotype, and
papain, ficin, bromelin, pronase, or chymotrypsin, but trypsin does not presence on Fy(a – b – ) cells from people of non - African origin.
abolish Fyᵃ and Fyᵇ activity. Like Fy3, Fy5 is a protease – resistant antigen.
Fy5 is expressed equally strongly on red cells of adults and newborns
#MLSWATER2022 Page 11 of 18
MODULE 3: MINOR BLOOD SYSTEMS
DARC appears to be part of a protein complex that contains the Rh Duffy Glycoprotein
proteins, so Fy5 expression could be dependent on an interaction Binds a variety of pro- inflammatory chemokines and is often known
between those proteins. as the Duffy Antigen Receptor for Chemokines (DARC)
Duffy antigens are located on a glycoprotein of apparent MW 35 –
50 kDa, a chemokine receptor of the G protein - coupled family.
Duffy Antibodies Duffy glycoprotein is present on endothelial cells of post - capillary
venules and on other cells throughout the body
1. Anti – Fyᵃ
Individuals with the Fy(a – b – ) phenotype are resistant to infection
Anti - Fyᵃ is estimated to be three times less frequent than anti – K
by the malarial parasite Plasmodium vivax and Fy(a – b – ) red cells
Duffy antibodies are not commonly detected in the first 6 months
are refractory to invasion by P. vivax in vitro
following transfusion, but, relative to other antibodies, are more
Interaction between the Duffy glycoprotein and receptors on P.
common after 6 months and even more so after 5 years
vivax merozoites are essential, but not sufficient, for red cell
Anti – Fyᵃ are usually IgG, mostly IgG1
invasion.
Anti-Fyᵃ generally react best by an antiglobulin test, but rarely anti -
The Duffy (FY or DARC) locus is on chromosome 1q21 - q22.
Fyᵃ may be directly agglutinating.
o DARC is N - glycosylated, but has no, or very little, O -
Anti – Fyᵃ in donor blood was responsible for a transfusion reaction
glycosylation.
in a Fy(a + b + ) patient
Treatment of red cells with the N – glycanase endo F prior to
solubilization and immunoblotting with anti - Fyᵃ resulted in a
Anti - Fyᵇ dramatic reduction in apparent MW and sharpening of the band on
Anti - Fyᵇ is a relatively rare antibody usually found only in mixtures the blot
of red cell antibodies. Similar results were obtained by treatment of purified DARC or of
o It has been stimulated by pregnancy and transfusion, and by tryptic peptide derived from it
intrauterine transfusion in the mother Variation in the degree of N - glycosylation may account for the
o naturally occurring anti - Fyᵇ has been found range of MW
o Often consisting entirely of IgG1
o Fyᵇ antibodies generally react best by an antiglobulin test, but
Duffy Chemokines
directly agglutinating examples are known
Chemokines are chemotactic cytokines that are involved in many
o Some anti - Fyᵇ bind complement
cellular processes, especially the recruitment, activation, and
o Anti - Fyᵇ has been responsible for immediate and delayed HTRs,
directional movement of leucocytes
two of which were reported to have been fatal
There are two main classes of chemokines, called CXC and CC based
on the position of two highly conserved cysteine residues at their N
Anti - Fy3 - termini, plus two minor classes, C and CXXXC.
Anti - Fy3 is potentially hemolytic and has been responsible for o Most chemokine receptors belong to a very large family of
immediate and delayed HTRs, including intravascular hemolysis in integral cell - membrane glycoproteins, G protein - coupled
an acute reaction and hyper hemolysis in a patient with sickle cell receptors, which traverse the membrane seven times and
disease have an extracellular N - terminal domain
Fy(a−b−) red cells should be selected for transfusion to patients with o Most chemokine receptors are specific for one or more
anti - Fy3. chemokines of a single class, but DARC, a promiscuous
receptor, binds with high affinity to 60% of inflammatory

#MLSWATER2022 Page 12 of 18
MODULE 3: MINOR BLOOD SYSTEMS
chemokines from both CX and CC classes, but not with OTHER BLOOD GROUP ANTIGENS
homeostatic chemokines LUTHERAN BLOOD GROUP SYSTEM
o Unlike almost all other G protein - coupled receptors, DARC Two most common antigen (A and B)
lacks the Asp - Arg – Tyr (DRY) motif in the second The Lutheran system consists of 20 antigens: LU1 to LU22 in the
cytoplasmic loop and does not appear to be coupled to a numerical notation, with two declared obsolete
guanosine triphosphate - binding protein (G - protein) o Four pairs of these antigens have allelic relationships and
o Duffy is a chemokine - binding protein with no signalling represent SNPs in the Lutheran gene, LU :
function and has been referred to as a ‘ silent receptor ’ or • Luᵃ (LU1) and Luᵇ (LU2);
interceptor (internalising receptor)
• Lu6 and Lu9;
o DARC is present on the endothelial cells of post -capillary
• Lu8 and Lu14; and
venules throughout the body, in both Duffy -positive people
• Auᵃ (LU18) and Auᵇ(LU19)
and Fy(a – b – ) Africans
The null phenotype, Lu null or Lu(a – b – ), in which the red cells
Fy(a –b–) human red cells are refractory to invasion by P.
lack all Lutheran system antigens, results from homozygosity for
knowlesi and P. vivax merozoites, in vitro , whereas Fy(a+b+)
inactivating in the Lutheran gene
cells are invaded
o Individuals with the Lu null phenotype may make an
o Despite being Fy(b + ) and Fy:3, red cells of the Old World
antibody to the Lutheran glycoproteins (Lu - gps), anti - Lu3
rhesus monkey are Fy: – 6 and are not invaded by P. vivax ,
o Nucleotide changes in LU are associated with loss of eight
but are invaded by P. knowlesi
other antigens of high frequency, which are also absent
o New World capuchin monkey cells are Fy(a – b – ) Fy:3, – 6
from Lu null cells
and are not invaded by P. knowlesi or P. vivax merozoites
o The molecular bases for two other antigens of high
o These data suggest that the Fy6 epitope is important forthe
frequency absent for Lu null cells are unknown.
invasion of P. vivax
Lutheran antigens are located on two red cell membrane
o The Duffy gene is subject to opposing selection pressures:
glycoproteins (CD239) of apparent MW 78 and 85 kDa, which
the need to maintain effective chemokine binding versus
belong to the immunoglobulin superfamily of receptors and
the benefits of reducing P. vivax invasion.
adhesion molecules.
The Lu - gps are ligands for the extracellular matrix glycoprotein,
Disease associations laminin LU or BCAM is situated on chromosome 19q12 - q13 and
HIV - 1 attaches to red cells via DARC, effecting infection of target consists of 15 exons, with alternative splicing accounting for the two
lymphocytes isoforms of the Lu – gps
Red cell Duffy expression is strongly associated with severity of
sickle cell disease (SCD) and especially with organ damage.
The Lutheran glycoproteinsis Lu –gps
o Twice as many Duffy - negative patients had evidence of
Components of apparent MW 85 and 78 kDa were revealed by
organ damage compared with Duffy -positive patients;
immunoblotting of red cell membranes with monoclonal anti - Lu b
o Duffy - negative patients were nearly four times more likely
or with alloanti - Luᵃ , - Luᵇ, - Lu3, - Lu4, - Lu6, - Lu8, - Lu12, - Auᵃ, or
to have proteinuria
- Auᵇ
o Duffy - positive SCD patients have higher plasma levels of
chemokines IL - 8 and RANTES than Duffy - negative patient
The Lu-glycoproteins belong to the immunoglobulin superfamily (IgSF)
The immunoglobulin superfamily (IgSF) is a large collection of
glycoproteins, abundant on leucocytes, but also present on other
#MLSWATER2022 Page 13 of 18
MODULE 3: MINOR BLOOD SYSTEMS
cells, which contain repeating extracellular domains with sequence Lutheran Antibodies
homology to immunoglobulin variable (V), constant (C1 or C2), or Anti- Luᵃ
intermediate (I) domains. Luᵃ antibodies are usually IgM, but, like other Lutheran - system
o Each IgSF domain consists of approximately 100 amino acids antibodies, often have IgG and IgA components
and is structured into two β – sheets stabilised by a Anti - Luᵃ often agglutinate Lu(a + ) red cells directly, with a thermal
conserved disulphide bond optimum well below 37 °с
o IgSF glycoproteins mostly function as receptors and o Some also react in an antiglobulin test, and a few,
adhesion molecules, and may be involved in signal predominantly IgG examples, are reactive only by an
transduction antiglobulin test
o A single - chain variable - fragment (scFv) with Luᵃ specificity
Luᵃ and Luᵇ (Lu1 ad Lu2) has been produced by phage display and recombinant DNA
The first Lutheran antibody, anti - Luᵃ, was described in 1945 by technology, and a monoclonal anti - Luᵃ constructed
Callender et al. Anti-Luᵇ
Anti-Luᵇ is relatively rare, often found as a single antibody
Variation in Antigenic Strength o It has been stimulated by transfusion and by pregnancy
The Lutheran antigens are very variable in strength o Anti - Luᵇ are often optimally active in the antiglobulin test,
o Luᵃ on red cells from different families may vary but directly agglutinating anti - Luᵇ have been described,
quantitatively, but the antigenic strength remains roughly many with a temperature optimum of about 20°с.
constant within the family. o Most anti - Luᵇ are mixtures of IgG and IgM, although IgA
o Occasionally adsorption and elution tests are required to may also be present
detect weak Luᵇ on Lu(a + b + ) cells o IgG anti - Luᵇ may be predominantly IgG1, although IgG2
o There is also heterogeneity of Lutheran antigen strength and IgG4 may be present
between individual red cells within a person, which
accounts for the characteristic mixed - field agglutination
patterns often seen with Lutheran antisera, especially anti -
Luᵃ, and the wide range of survival times of Lu(b + ) cells
introduced into an Lu(a + b – ) person with anti - Luᵇ
o The abundance of Luᵇ antigens on red cells, as determined
by Scatchard analysis with purified monoclonal anti - Luᵇ, is
relatively low and shows wide variation
Red cells from cord samples and from infants in the first year of life
have markedly weakened expression of Luᵃ and Luᵇ compared with
those from adults
o Adult levels of Luᵃ and Luᵇ antigenic expression are reached
by the age of 15

#MLSWATER2022 Page 14 of 18
MODULE 3: MINOR BLOOD SYSTEMS
Clinical Significance of anti - Luᵃ and - Luᵇ Recombinant Lutheran Antigens
No case of HDFN caused by anti - Luᵃ or - Luᵇ and requiring any Lutheran antigens have been used as models for the application of
treatment other than phototherapy is reported, although raised recombinant proteins in antibody identification
bilirubin or a positive DAT may be detected Recombinant proteins containing all or some of the IgSF domains of
No case of HDFN caused by anti - Luᵃ or - Luᵇ and requiring any the Lutheran protein have been expressed in eukaryote or
treatment other than phototherapy is reported, although raised prokaryote cells
bilirubin or a positive DAT may be detected: The purified protein was then used in agglutination inhibition tests,
o Babies of mothers with high - titre IgG1 anti - Luᵇ or - attached to polystyrene plates for detection by an ELISA procedure,
Lu6 had no sign of HDFN, their red cells gave negative or coupled to superparamagnetic particles for detection in a particle
DATs, and Lutheran antibody could not be detected in gel immunoassay.
their sera. Alloanti-Luᵃ or Alloanti-Luᵇ were detected with high sensitivity and
o Maternal IgG1 usually becomes concentrated in the specificity
fetal circulation by active placental transfer.
o As Lu-gp is present on placental tissue, it is possible that Effects of Enzymes and Reducing Agents on Lutheran Antigens
Lutheran antibodies are adsorbed by placental cells, Lutheran antigens are destroyed by treatment of red cells with
preventing their transfer to the fetus trypsin or α - chymotrypsin; papain has little effect
Sulphydryl reducing agents, such as AET and DTT, break inter- and
intra - polypeptide chain disulphide bonds resulting in the unfolding
Other Lutheran antigens and antibodies of the protein
All Lutheran antigens are absent from Lu null cells and absent from Red cells treated with 6% AET or 200mM DTT at pH8.0 did not react
or expressed very weakly on In(Lu) cells with most Lutheran antibodies tested, including many examples of
anti - Luᵃ and - Luᵇ
LU11
Lu11 has not been shown to be inherited and has not been shown
to be located on the Lu-gps or encoded by the LU gene, and so Lu null and Anti-Lu3 (LU3)
should be referred to as a para -Lutheran antigen Lu null phenotype is extremely rare and has a recessive mode of
inheritance
LU6 and LU9
o Lu null cells lack all Lutheran system antigens
Lu6 and Lu9, Lutheran antigens of high and low frequency,
o Individuals with the Lu null phenotype may make an
respectively, have an antithetical relationship, and represent a SNP
antibody, anti - Lu3, which reacts with all red cells apart
and Cfo I restriction polymorphism in LU encoding an amino acid
from those with the Lu null phenotype
substitution in the third IgSF domain
o Lu null red cells have normal expression of those antigens,
such as AnWj, that are expressed very weakly on In(Lu) red
LU8 and LU14 cells
Lu8 and Lu14, Lutheran antigens of high and low frequency,
respectively, have an antithetical relationship, and represent a SNP
Anti-Lu3 (-LU3)
and Fat I and Nla III restriction polymorphisms in LU , encoding an
All Lu null propositi have been found following the detection of an
amino acid substitution in the second IgSF domain
antibody to a high frequency antigen, anti-Lu3
o Anti - Lu3 has a single specificity and reacts equally strongly
with Lu(a+b–), Lu(a + b + ), and Lu(a – b + ) cells
#MLSWATER2022 Page 15 of 18
MODULE 3: MINOR BLOOD SYSTEMS
o Adsorption with Lu(a + b – ) cells will remove the activity for For these reasons, the presence of Lewis antibodies in a patient’s
Lu(a – b + ) cells and vice versa [90,92] serum does not require transfusions of Lewis negative RBCs, as long
o Lu3 is present on all red cells that express any Lutheran as pre-transfusion tests performed at 37⁰C and Coombs phase are
antigen compatible and there is no evidence of in vitro hemolysis.
Luᵇ was not detected on lymphocytes, granulocytes, monocytes, The Lewis antibodies reactive at 37⁰C and antihuman globulin
platelets, or the erythroleukaemic cell lines K562 and HEL phase, however, should not be ignored because these antibodies
can cause in vivo RBC destruction. In the presence of multiple
Disease associations antibodies, Lewis antibodies often complicate antibody
The IgSF gps expressing Lutheran and LW blood group activity are identification, but they can be easily inhibited with saliva from
overexpressed on SS red cells in sickle cell disease: SS red cells secretors or with commercially available Lewis substance.
express about 67% more Lu-gp than normal cells and bind increased
quantities of laminin Factors Contributing to Clinical Insignificance of Lewis Antibodies
o Neutralization of Lewis antibodies by Lewis substances present in
For more information regarding Lutheran Blood Group and probably the plasma
everything that confuses you. Refer to: Daniels, Human Blood Group, 3rd o Loss of red cell Lewis antigen(s) into the plasma
Edition https://books.google.com.ph/books?id=CHT_- o Lack of reactivity at 37⁰C and antihuman globulin phase
c45qQcC&printsec=frontcover o Generally, lgM in nature and incapable of crossing placenta
o Lewis antigens poorly developed in newborn infants

CLINICAL SIGNIFICANCE OF ANTIBODIES OF MINOR BLOOD The MNS (002) Blood Group System
GROUPS Disease Association
Lewis Blood Group GPAM may serve as the receptor by which certain
Although some cases of hemolytic transfusion reactions caused by pyelonephritogenic strains of Escherichia coli gain entry to the
anti-Lea have been reported and there have been cases of in vivo urinary tract.
RBC destruction due to anti-Leb, Lewis antibodies are generally The malaria parasite Plasmodium falciparum appears to use
considered insignificant in blood transfusion practices. This is alternative receptors, including GPA, GPB, and GPC23 for cell
because: invasion; some of these receptors also involve NeuNAc.
1. Lewis antibodies can be neutralized by the Lewis substances In an attempt to identify the receptor, the invasion rate into cells
present in the plasma and can thereby be decreased in quantity. with normal and rare phenotypes was studied. Reduced invasion is
2. The Lewis antigens dissociate from the RBCs as readily as they seen with En(a), U, MkMk, Tn and Cad RBCs (which have altered
bind to the RBCs. In other words, the Lewis-positive donor RBCs oligosaccharides on glycophorins), Ge, and normal RBCs treated
can become Lewis-negative RBCs following transfusion into an with neuraminidase and trypsin
individual with a Lewis-negative phenotype. These antigens
released into the plasma can further neutralize any Lewis
antibodies present in the recipient plasma.
3. Lewis antibodies are generally IgM and therefore cannot cross
the placenta and cause HDN. In addition, Lewis antigens are not
fully developed at birth.

#MLSWATER2022 Page 16 of 18
MODULE 3: MINOR BLOOD SYSTEMS
The P Blood Group: P (003) and Globoside (028) Blood Group Systems lymphocytic leukemia have less lymphocyte i antigen than normal
and Related Antigens control subjects.
Disease Associations Chronic dyserythropoietic anemia type II or hereditary
Several pathologic conditions associated with the P blood group erythroblastic multinuclearity with a positive acidified serum test
antigens have been described: parasitic infections are associated (HEMPAS) is associated with much greater i activity on RBCs than
with anti-P1 , early abortions with anti-PP1 Pk or anti-P, and PCH control cord RBCs.
with autoanti-P. The P system antigens may also be associated with HEMPAS RBCs are very susceptible to lysis with both anti-i and anti-
urinary tract infections. I, and lysis by anti-I appears to be the result of increased antibody
Some pyelonephritogenic strains of E. coli ascend the urinary tract uptake and increased sensitivity to complement.
in ladderlike fashion by adhering to P1 and/or Pk glycolipids on I antigen may be involved in binding immune complexes consisting
uroepithelial cells. The fimbriae or pili of such organisms have of drug and drug antibodies. Immune complexes involving rifampin,
receptor sites for structures involving Gal(ß1-4)Gal( 1-4), the nitrofurantoin, dexchlorphenyramine, and thiopental have been
terminal sugars for P1 and Pk. associated with binding to I antigens on RBCs and subsequent
Other globoside associations have been identified with infection. complement activation and hemolysis.
Streptococcus suis, which occasionally causes meningitis and In Asians, the adult i phenotype has been associated with congenital
septicemia in humans, binds exclusively to Pk antigen. A class of cataracts. Mutations at the I locus have been identified in three
toxins secreted by Shigella dysenteriae, Vibrio cholerae, Vibrio Taiwanese families with the adult i phenotype, which suggests a
parahaemolyticus, and some pathogenic strains of E. coli also have molecular genetic mechanism for this condition in this population.
binding specificity for a Gal(ß1-4)Gal( 1-4) moiety. In addition,
globoside is the receptor of human parvovirus B19. The Kell (006) and Kx (019) Blood Group Systems
Antibodies of the Kell system should be considered potentially
The I (027) Blood Group System and i Antigen clinically significant, both from the point of view of causing severe
Disease Associations hemolytic disease of the fetus and newborn (HDFN) and hemolytic
Anti-I and cold agglutinin syndrome and M. pneumoniae transfusion reactions (HTRs).
Patients with Kell-system antibodies should be transfused with
Anti-i and infectious mononucleosis
Anti-IT and Hodgkin’s lymphoma. antigen-negative blood whenever possible.
Cold autoantibodies have also been reported in influenza infections, Kell-system antibodies are usually IgG and predominantly IgG1.
but other associations are rare. HDFN caused by anti-K differs from that resulting from anti-D.
Diseases can also alter the expression of I and i antigens on RBCs. Anti-K HDFN is associated with lower concentrations of amniotic
Conditions associated with increased i antigen on RBCs include fluid bilirubin than anti-D HDFN of comparable severity. Postnatal
those with shortened marrow maturation time or dyserythropoiesis: hyperbilirubinemia is not prominent in babies with anemia caused
acute leukemia, hypoplastic anemia, megaloblastic anemia, by anti-K. There is also reduced reticulocytosis and erythroblastosis
sideroblastic anemia, thalassemia, sickle cell disease, paroxysmal in the anti-K disease, compared with anti-D HDFN
The infrequent cases of HDN caused by Kell immunization tend to
nocturnal hemoglobinuria (PNH), and chronic hemolytic anemia.
result in severe fetal anemia because maternal anti-Kell target fetal
Except in some cases of leukemia, the increase in i on RBCs is not
usually associated with a decrease in I antigen; the expression of I red blood cell (RBC) precursors, suppressing the fetal production of
antigen can appear normal or sometimes enhanced. RBCs.
Reactive lymphocytes in infectious mononucleosis are reported to
have increased i antigen. Those from patients with chronic
#MLSWATER2022 Page 17 of 18
MODULE 3: MINOR BLOOD SYSTEMS
The Duffy (008) Blood Group System
The Duffy-Malaria Association
Since 1955, Miller and colleagues confirmed that malaria
merozoites invaded only RBCs carrying normal Fya or Fyb antigen.
When antigen sites were blocked by antibody or denatured with
certain enzymes, the RBCs became resistant to invasion. Because
the resistance factors for P. knowlesi and P. vivax were so parallel in
West African populations, it was suggested that Fya and Fyb might
also be the invasion receptor for P. vivax.
Close evaluation of the invasion process of P. knowlesi suggests that
two receptor sites are involved: one for attachment and one for
invasion.Initial attachment of the merozoite to the RBC occurs
regardless of Duffy type, but the junction and invasion are Duffy
antigen–dependent.
Data from human and old and new world monkey RBCs and their
susceptibility to invasion by P. vivax and P. knowlesi indicate that
Fy6 is important for invasion for P. vivax. The monoclonal anti-Fy6
has been shown to block invasion of RBCs by P. vivax

The Kidd (009) Blood Group System


Disease Associations
Although Jka and Jkb are thought to be human RBC antigens, three
organisms have been associated with Jkb-like specificity. Two,
Enterococcus faecium and Micrococcus, were able to convert Jk(b)
cells to Jk(b), and one, Proteus mirabilis, may have been the
stimulus for an autoanti-Jk

-END OF TRANSCRIPTION -

#MLSWATER2022 Page 18 of 18
MODULE 4: PRE-TRANSFUSION TESTING
MLS 18, March 17, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
The crossmatch is an actual mixing of the recipient and donor’s blood to
Legend: insure in vitro compatibility.
Transcription Bullet Only a part of compatibility testing
Notes/Module Packet Bullet Check for any untoward reaction between the donor and recipient’s
 PPT blood
MODULE OUTLINE
I. Compatibility Testing SEROLOGIC CROSSMATCH
II. Antibody Screening Two main functions of the serologic crossmatch test:
III. Selection of Appropriate Donor Units o It is a final check of ABO compatibility between donor and patient.
IV. Routine Blood Bank Laboratory o It may detect the presence of an antibody in the patient’s serum
that will react with antigens on the donor RBCs but that was not
LEARNING OUTCOMES detected in antibody screening because the corresponding antigen
1. Define compatibility test. was lacking from the screening cells.
2. Identify tests included in compatibility test. Consists of mixing the patient’s serum with the donor’s RBC
3. Describe appropriate methods for proper patient identification in
sample collection.
Major Crossmatch
4. Outline the procedure for testing donor and patient specimens.
An in-vitro determination that combines recipient’s plasma with a
5. Compare and contrast major and minor crossmatch procedures.
5% cell suspension of the donor’s cells.
6. Resolve incompatibilities in the crossmatch.
This test is performed at different phases in the same manner as the
7. State the limitations of compatibility testing procedures.
antibody screen and antibody identification.
8. Discuss future issues of compatibility testing.
The immediate spin crossmatch is the first phase of the major
crossmatch procedure.
o This procedure provides a determination of compatibility
COMPATIBILITY TESTING
that encompasses ABO incompatibility.
Series of procedures designed to ensure the safety of blood for
The final phase is the anti-human globulin (AHG) test.
transfusion.
o Antibodies present in the recipient that are reactive at the
Vital part of the blood bank laboratory. AHG phase are most likely to cause an in vivo reaction
o The determination of compatibility of a unit of blood for transfusion
is performed each time a unit of red blood cells is to be transfused.
Minor Crossmatch
Included in the compatibility test is a crossmatch.
Performed by mixing donor plasma with cells from the recipient.
The minor crossmatch is no longer performed.
CROSSMATCHING/CROSSMATCH TESTING The majority of cellular products transfused are red blood cells
The crossmatch test is the testing of the patient’s serum with the donor rather than whole blood.
RBCs, including an antiglobulin phase or simply an immediate spin phase
to confirm ABO compatibility.
The volume of donor plasma transfused with red cell products is Adding ethylenediaminetetraacetic to the test system reportedly
very small and not significant enough to induce a reaction with eliminates some of the false-positive reactions, thus improving the
recipient cells sensitivity of the immediate spin crossmatch.
2-3 drops of serum sample with a drop of a wash of 2-5% RBC
Compatibility test results: suspension; observe presence of hemolysis in the centrifuge
Compatibility test procedures are performed and the results accomplished by mixing the recipient’s serum with the donor’s RBC
assessed. and centrifuging immediately (no incubation)
Compatible units of red cells will have no agglutination or hemolysis
at any phase of testing. Type-And-Screen Procedure
Tests that have either hemagglutination or hemolysis noted are Involves testing the patient’s blood sample for ABO, Rh, and
incompatible and should not be transfused without further clinically significant unexpected antibodies.
evaluation. The patient sample is then stored in the blood bank refrigerator for
The cause of the incompatibility should be determined and future crossmatch if blood is needed for transfusion.
documented. The type-and-screen procedure has application for patients
undergoing many elective procedures who may need blood;
because the process may not always require transfusion, the
Objective of testing: to select donor units that can provide maximal crossmatch is not performed until necessary.
benefit to the patient, which should be kept in mind when developing
the test protocol.
37°C Incubation with the use of enhancement media:
Enhancement Media – there is the option to use 22% albumin, low
Conventional Crossmatching ionic saline solution, or polyethylene glycol
Three Phases: Immediate Spin Crossmatch, 37°C Incubation with
the use of enhancement media, Antiglobulin Crossmatch Antiglobulin Crossmatch
The antiglobulin crossmatch procedure begins in the same manner
Immediate Spin Crossmatch as the immediate spin crossmatch, continues to a 37°C incubation,
This is accomplished by mixing the recipient’s serum with donor and finishes with an antiglobulin test.
RBCs and centrifuging immediately. Several enhancement media may be applied to boost antigen-
Absence of hemolysis or agglutination indicates ABO compatibility. antibody reactions
The immediate spin does not detect all ABO incompatibilities o These may include albumin, low ionic strength solution
False reactions may be seen in: (LISS), polyethylene glycol, and polybrene
o The presence of other immediate spin-reactive antibodies For greatest sensitivity, an antihuman globulin (AHG) reagent
(e.g., autoanti-I) containing both anti-IgG and anticomplement may be selected for
o Patients with hyperimmune ABO antibodies the final phase of this crossmatch method.
o May be seen when the procedure is not performed correctly An autocontrol, consisting of the patient’s own cells and serum, may
(e.g., delay in centrifugation or reading) be tested in parallel with the crossmatch test.
o When rouleaux is observed Used to detect (unagglutinated IgG) antibodies that were not
o When infants’ specimens are tested. detected with 37°C Incubation and Immediate spin crossmatch

#MLSWATER2022 Page 2 of 16
Computer Crossmatch Blood Sample Collection and Labelling
Uses computer technology that is developed and increases
sensitivity of tests Acceptable collection tubes for pre-transfusion testing are:
As computer technology has developed and the sensitivity of  Plain Red Tube (No Anticoagulant; No Serum Separator)
antibody detection has increased, a process for computer or  Yellow Top Tube (Acid Citrate Dextrose or ACD, Formula B)
electronic crossmatch has been developed.  Pink or Purple Top Tube (EDTA)
The electronic crossmatch is a viable option when there is no  Blue Top Tube (Sodium Citrate)
current or previous history of clinically significant antibodies.
One of the primary advantages is maximum utilization of blood Historically, the use of anticoagulant for crossmatching was
supply. discouraged, since the binding of calcium inhibited the activation of
When the electronic crossmatch is used with appropriate patients, complement in the compatibility test.
wastage of units by holding units for specific patients is significantly o The clinical significance of these complement-binding
reduced. antibodies has been determined insignificant. Hence, the
Many believe that the computer crossmatch is safer than the use of anticoagulated samples has become acceptable.
immediate spin because of the integrity of the computer software Anticoagulated samples are readily available for crossmatching in an
to detect ABO incompatibility between the sample submitted for emergency situation since the time required for a sample to clot
pretransfusion testing and the donor unit. prior to processing may delay testing and treatment.
The computer crossmatch compares recent ABO serologic results o In addition, the formation of fibrin that may interfere with
and interpretations on file for both the donor and the recipient testing is less likely when using an anticoagulated specimen.
being matched and determines compatibility based on this The sample should not be contaminated with IV solutions
comparison. o A sample may be collected from an infusion line only if the
Advantages of the Electronic Crossmatch: line is flushed with saline and a volume of blood equal to
a. More efficient management of blood inventory twice the test volume is withdrawn and discarded prior to
b. Less wastage of blood components due to outdate obtaining the sample.
c. More efficient use of technician time The collection process should employ a needle of sufficient size to
d. Increase in staffing flexibility avoid hemolyzing the red cells in the sample.
e. Smaller recipient sample required o The presence of hemolysis in plasma of the original sample
f. Potential for a centralized transfusion service data bank may mask hemolysis resulting from an antigen-antibody
accessible by all interested parties interaction in the testing process.
g. annual savings Lipemic serum may also provide an environment that is difficult for
h. Reduced sample requirements evaluation of serological reactions.
Reduced expenses o At times it may be necessary to accept test samples with
i. Reduced handling of biological materials some amount of hemolysis or lipemia.
j. Elimination of false reactions associated with the immediate o Each institution should establish criteria and procedures for
spin crossmatch. acceptable samples
Individual institutions should establish the maximum age for pre-
transfusion testing samples.

#MLSWATER2022 Page 3 of 16
Patient specimen free transfusion testing begins with properly These two independent identifiers should be confirmed at the
labelled, properly collected and identified patient specimen; patient time of collection and the blood sample labeled with this unique
identification is first in compatibility testing information:
Proper identification of the patient is imperative; patient must have the date of collection
a wristband with the identification information, tube should be The phlebotomist’s signature.
labelled on the bedside immediately after the sample was drawn This labeling must take place at the bedside.
Label should include the date, time of collection, name of the
person collected, and sample should not be collected into a Most institutions use hospital identification bands to identify
previously labeled tube. patients
During collection serum is a preferred specimen for compatibility o Past records should be compared to current test results.
testing, hemolysis should be avoided, and blood should not be The requirement further states that the current ABO and Rh
drawn from intravenous site. type must be compared to results obtained within the past
Stop the infusion for 5-10 minutes before collecting blood and then 12 months.
the lye should be flush with normal saline and first 5-10ml blood lye o Discrepancies are investigated and resolved before any
should be discarded. units are issued for transfusion.
o Red top for serum sample The AABB Standards require that all donor blood have the ABO and
o Yellow, pink and purple top used for collecting from donor Rh confirmed from an attached segment prior to transfusion of any
Fresh sample (not older than 72 hours) should be the best option whole blood or red blood cell component.
used for compatibility testing if patient has previously undergone o The standards do not require that the institution issuing the
transfusion or transfusion history is unknown. blood for transfusion perform a test for the confirmation of
o Also, the same for pregnant women the weak D antigen
For sample storage, EED standards states that patient sample According to the AABB, patient samples must be tested for ABO, Rh,
should be stored between 1° C and 6° Celsius for at least 7 days and unexpected antibodies to red cell antigens prior to transfusion
following transfusion. of red cells.
Lipemic samples or lipemia can interfere with the testing o The weak D test is not required, but the antibody screen
Hemolyzed sample should also be avoided must include “incubation at 37°C preceding an antiglobulin
test using reagent red cells that are not pooled.”
AABB Standards for Blood Bank and Transfusion Services, 25th Edition, The compatibility test procedure is only applicable for red cell
states that a recipient sample must be: products that are to be transfused
a. Collected within three (3) days of the scheduled transfusion if o Plasma based products such as cryoprecipitate, platelet
the patient has been transfused or pregnant in the preceding concentrates, and frozen plasma contain virtually no red
three (3) months or if the history is uncertain or unavailable. cells and require no compatibility testing.
The day of collection is labeled as day zero (0). o These components have been screened for antibodies and
b. These patient samples, as well as a segment from any red cell infectious diseases. Hence, they are transfused as ABO-
containing component, are all refrigerated and retained for at compatible.
least seven (7) days after transfusion. o Platelet-pheresis products and granulocyte concentrates
may contain some red cells, but require crossmatching only
The AABB designates that the request for blood and blood products if more than 2 ml of red cells are present.
contain sufficient information to uniquely identify the patient. This
includes a minimum of two independent identifiers:
#MLSWATER2022 Page 4 of 16
PROBLEM SOLVING INCOMPATIBLE CROSSMATCHES o The procedure must include a system to ascertain that the
autologous units are administered before any allogeneic units
PROBLEM are issued.
POSSIBLE CAUSE RESOLUTION o The administration of allogeneic units before the stored
ENCOUNTERED
ABO typing  Patient identification  Repeat the ABO autologous units presents a liability risk for the administering
error group of the institution.
 Sample identification recipient
error  Recheck the label COMPATIBILITY TESTING OF NEONATES UNDER 4 MONTHS OF AGE
 Choice of unit of of the selected unit Neonates require special consideration
incompatible ABO  Recollect a sample Antibodies present have originated from the mother since the
group from the recipient neonate is unable to produce their own antibodies.
Unexpected  Cold alloantibody  Test with panel Neonates actually have the antibody of their mother
antibodies when  Cold autoantibody cells and Initial pretransfusion testing uses the neonate’s red cells for ABO
antibody screen is  anti-A1 in an A2 determine clinical and Rh typing.
negative at AHG individual significance of Reverse grouping is not performed on neonates due to the lack of
antibody antibody production.
 Prewarm Antibody screen may be performed using either the infant or
compatibility maternal plasma.
testing If the initial antibody screen is negative, there is no need to perform
 Test plasma of additional antibody screens or crossmatches on the neonate during
recipient with A2 this admission, so long as ABO-compatible and Rh identical red cell
cells products are administered.
Positive AHG phase  If the antibody screen is positive, the antibody must be identified
Positive DAT in donor  Perform DAT test
and antigen negative units administered for assured compatibility.
of compatibility  Antibody to low on donor
Additionally, an antiglobulin crossmatch should be performed and
test incidence antigen in  Choose alternate
only compatible units released.
recipient with antigen unit for
Crossmatching of all units needs to be continued until the maternal
positive cells in donor compatibility test
antibody is no longer detectable in the infant’s plasma.
Units of blood are crossmatched and divided since only small
COMPATIBILITY TESTING FOR AUTOLOGOUS TRANSFUSION quantities are administered to neonates.
Autologous donation is the process of collecting units of blood and Blood bags with multiple satellite bags or “pedi-packs” are
storing them for transfusion to the original donor commercially available for this purpose.
o This procedure is most often used for patients anticipating o This further reduces the possibility of incompatibility or
elective surgery. disease transmission
o Procedures to ensure that the collected units are transfused CMV negative units for use specifically in neonates may be obtained
to the intended recipient are established by the transfusing from the component source
institution.
o This procedure may be computerized with all records retained
electronically.

#MLSWATER2022 Page 5 of 16
CAUSES OF POSITIVE RESULTS IN THE SEROLOGIC CROSSMATCH Goal of Antibody Screening
Incorrect ABO grouping of the patient or donor. o To detect as many clinically significant antibodies as
An alloantibody in the patient’s serum reacting with the possible
corresponding antigen on donor RBCs. o To detect as few insignificant
An autoantibody in the patient’s serum reacting with the o To detect the red cell antibody other than anti a and anti b
corresponding antigen on donor RBCs.
Prior coating of the donor RBCs with protein, resulting in a positive SELECTION OF APPROPRIATE DONOR UNITS
antihuman globulin test. The first choice for transfusion is blood and blood components of
Abnormalities in the patient’s serum. the patient’s own ABO and Rh group. This is defined as ABO group–
Contaminants in the test system specific.
When blood and blood components of the patient’s ABO blood
ANTIBODY SCREEN group are not available or some other reason precludes their use,
The recipient’s serum or plasma must be tested for clinically units selected must lack any antigen against which the recipient has
significant unexpected antibodies. a clinically significant antibody.
Usually done or is part of the antibody testing When a recipient must be given blood of a different ABO group,
The object of the antibody screening test is to detect as many only packed RBCs can be given.
clinically significant unexpected antibodies as possible. Whole blood cannot be administered in these situations because
In general, “clinically significant unexpected antibody” refers to incompatible, preformed ABO antibodies are present in the whole-
antibodies that are reactive at 37°C or in the antihuman globulin blood plasma.
test and are known to have caused a transfusion reaction or o Ex. Group A whole blood cannot be transfused into a group
unacceptably short survival of transfused RBCs AB recipient, because the plasma of the group A whole
Detection of unexpected antibodies is important for the selection of blood has anti-B antibodies present.
donor RBCs that will have the best survival rate in the patient’s Group O packed RBCs can be safely used for all patients; however,
circulation and reduce the risk of hemolytic transfusion reaction. conservation of a limited supply of group O blood should dictate its
Antibody screening or antibody panels are used use for recipients of other ABO types only in special circumstances.
If ABO group–specific blood is not available or is in low supply,
Antibody screening tests should demonstrate the presence of all
alternative blood groups are chosen
potentially clinically significant alloantibodies in the recipient’s
Rh-negative blood can be given to Rh-positive patients; however,
serum or plasma and indicate the need for further studies.
good inventory management should conserve this limited resource
All antibodies encountered in the screening test must be identified
for use in Rh-negative recipients.
to determine potential clinical significance and to decide whether
o But if the Rh-negative unit is near expiration, the unit
there is a need to select antigen-negative units for transfusion.
should be given rather than wasted.
Multiple antibodies are more commonly found in patients older
Rh-positive blood should not be given to Rh-negative female
than 60 years old or have undergone transfusion multiple times
patients of childbearing age.
are usually the ones detected with unexpected reactions
Transfusion of Rh-negative male patients and female patients
In antibody identification we make use of screening cells (cells that
beyond menopause with Rh-positive blood is acceptable as long as
have no antigen present)
no preformed anti-D is demonstrable in their sera.
3 Phases Involved (Immediate screen, 37°C, Antihuman globulin
phase)

#MLSWATER2022 Page 6 of 16
BLOOD BANK LABORATORY Donor Processing
A facility involved in the collection, storage, processing, and  Before unit blood can be placed in general inventory, testing must
distribution of human blood and blood products for transfusion be performed
 Blood Processing centers utilize the testing methods that will
BLOOD BANK AREA AND FUNCTION provide the safest blood products for patients
AREA FUNCTIONS Product Labelling
Component Preparation  Separation of whole blood into packed Appropriate ABO and Rh type
And Storage RBCs, plasma, platelets and Expiration date
cryoprecipitate Stored at proper temperature
 Storage of blood products at
appropriate temperatures Issue of Blood Products
 Apheresis procedure The individual in the blood bank who will issue the blood product
Donor Processing Donor units tested for: inspects the unit for any abnormal appearance and verifies that all
 ABO and Rh required transfusion forms and labels are complete and that they
 Antibody Screen adequately identify the transfusion recipient.
 Serologic Test for Syphilis
 Transfusion-Transmitted viruses Personnel Requirements
Product Labelling  RBCs and any other component are Clinical Laboratory Improvement Amendments of 1988 (CLIA ’88)
labeled establishes personnel qualifications for laboratories performing
 Products stored at their proper certain types of testing:
temperatures o include gel technologies
Main Laboratory Patient Samples tested for: o Solid phase RBC adherence.
o ABO and Rh
o Antibody Screen Standard Operating Procedures
o Crossmatch These manuals, usually located at the workbench and accessible to
o DAT all personnel, contain information outlining the operations of the
o Prenatal Evaluation laboratory; details on how, when and why particular activities are
o Postpartum Evaluation done; and procedures for all tests performed.
o Cord blood studies SOP manuals are integral components of any blood bank
o Issue blood products laboratory’s quality assurance program.
Reference Laboratory Resolution of: They are reviewed at least annually and updated on a regular basis
o ABO and Rh discrepancy to reflect changes in operations and implementation of new
o Antibody identification regulations.
o Positive DAT
o Warm autoantibodies
o Cold autoantibodies
o Transfusion reaction

#MLSWATER2022 Page 7 of 16
Equipment o In addition to patient information, the label should also
 Blood Storage Refrigerator include the date and time of collection and the name of the
 Donor Couches person who collected the sample
 Dielectric Tube Sealer 2. Collection
 Blood Mixer & Collector o Serum is the preferred specimen for compatibility testing.
 Platelet Incubator o Hemolysis should be avoided.
 Platelet Agitator o Blood should not be drawn from an intravenous site unless
 Plasma Expressor absolutely necessary. In such a case, the infusion should be
 ELISA Reader with washer stopped, the line should be flushed with normal saline, and
 Refrigerated Centrifuge the first 5 to 10 mL of blood should be discarded before the
 Binocular Microscope CH 21 specimen is collected.
 Universal Hot air oven 3. Age of specimen
 Bacteriological Incubation o The freshest sample possible should be used for compatibility
 Centrifuge Machine testing.
 Rh view box o If the patient has previously undergone transfusion or if the
transfusion history is unknown, the sample should be no older
than 72 hours.
SUPPLEMENTARY NOTES: PRE TRANSFUSION TEST ING o Pregnant patients should also be tested with samples not
CROSSMATCHING more than 72 hours old
4. Sample storage
Only a part of compatibility testing.
o The AABB requires that patient samples must be stored
Type and Screen procedure has emerged as an acceptable
between 1◦C and 6◦C for at least 7 days following
alternative to crossmatching blood
transfusion.
Major Crossmatch
COMPATIBILITY TESTING:
o DONOR RBCS + PATIENT SERUM
Minor Crossmatch for homologous transfusion (ALLOGENIC):
o DONOR SERUM + PATIENTS RBCS ABO and Rh on donor units
Autocontrol ABO and Rh on recipient
o PATIENT RBCS + PATIENT SERUM Antibody screening of recipient
Antibody identification
Autocontrol
SPECIMEN COLLECTION Crossmatch
Pretransfusion testing begins with a properly collected and
identified patient specimen for autologous transfusion:
ABO and Rh on autologous units
1. Patient identification ABO and Rh on recipient
o The tube should be labelled at bedside immediately after the Antibody screening and major crossmatch
sample is drawn o not required, although an immediate spin major crossmatch
is often performed
#MLSWATER2022 Page 8 of 16
for neonatal transfusion: FDA Licensed Screening Cells
ABO and Rh on the infant unspooled reagent red cells
Antibody screen on the infant or the mother 2-3 vials (R1R1, R2R2,rr)
o If the antibody screen is negative, a crossmatch is not detects clinically significant Ab
necessary reagents red cell express the ff: D,C,E, c,e
o If the donor cells are not group O, the infant must be tested M,N,S,s,P1,Lea,Leb,K,k,Fya,Fyb,Jka,Jkb
for anti-A and anti-B antibodies. If either is present, ABO- Autocontol not required
compatible RBCs should be used. A crossmatch is not DAT-not required
necessary
NOTE: Group O cells are used so that anti-A and anti-B will not interfere
ANTIBODY DETECTION in the detection of antibodies to other blood group systems.
Antibody detection/ antibody screening is testing the patient’s
serum against 2 or 3 reagent group O screening cells. ANTIGRAM/ WORKSHEET
Screening cells are commercially prepared cells suspension from It is a listing of the antigen make up of each screening cells that is
individual donor that is phenotyped for most common antigens. provided with every lot of screening cells.
In test tube testing: 3 phases Can be used as a worksheet during antibody detection process.
o Immediate spin o Caution: Lot number must match against the screen cells in
o 37C incubation use
o AHG Sample (link to worksheet):
Testing can also be: gel technique or solid phase method https://drive.google.com/drive/folders/1d_Lz_7fXfjYItsjN_yUhbbk7dStDOo
Performed as part of: Hi?usp=sharing
o testing to provide compatible red cell transfusion
o prenatal evaluation
o evaluation of HDFN ANTIBODY IDENTIFICAT ION
o resolution of transfusion reactions ANTIBODY PANEL
o processing of donor units
Just an expanded antibody screen
Uses group O reagent RBCs
Uses Antibody Panels - used to identify an unexpected antibody
RBCs from 8-20 donors
detected by the antibody screening
Patient serum or plasma
Expanded version of antibody screening
Autocontrol
antibody screening uses screening cells 2-3 reagents of group O
IS / 37 C / AHG if tubes
screening cells
AHG only if gel or solid phase
Antibody panel uses 8-20 screening cells, these panels usually
Reactions documented on a sheet that outlines every RBC’s
contain 10-15 or 8-20 vials of group O cells each of which yield a
phenotype
different antigen reaction pattern

Each of the panel cells has been antigen typed (shown on antigram)
o + refers to the presence of the antigen
o 0 refers to the absence of the antigen
#MLSWATER2022 Page 9 of 16
INTERPRETING ANTIBODY PANELS
1. Check History
Will give you clue what possible Ab is involved.
Check racial profile, recent transfusion, pregnancy and recent
bacterial or viral infection.
Notes the previous condition of the patient.
o Ex. pregnant: Rh blood group system is involved
Important also to note the racial profile of the patient since African-
American lacks Duffy antigen and Whites has high frequency
antigen

All cells are negative at AHG, so add “Check” Cells 2. Check Autocontrol
Check cells are added to all negative tubes. Negative - alloantibody
Check cells are cells coated with IgG and should react positively with Positive – autoantibody or DTR (i.e., alloantibodies)
the AHG in the tube.
o If check cells are negative, the procedure was not Check if the reaction if zero or plus or positive
performed correctly and should be repeated. If the reaction is positive that would indicate that there is presence
of autoantibody
If the reaction is negative there might be the presence of
alloantibody
o Another thing for negative control, it would show a uniform
reaction that means all cells are reactive. It would suggest there
might be presence of multiple alloantibodies and single
alloantibodies related with high frequency antigen
Antibodies will only react with cells that have the corresponding
antigen; antibodies will not react with cells that do not have the
antigen

3. Check the Reaction Phase


IS – cold reacting (IgM); clinically insignificant
37° - cold (some have higher thermal range) or warm reacting (IgG)
AHG – warm (IgG) and significant Ab. It detects Ab that have been
coated/sensitized RBCs but were not visible as agglutinates

4. Look at the general pattern (Reaction strength)


1 consistent strength (Uniform)– one antibody
Different strengths(Mixed reaction)s – multiple antibodies or
dosage

#MLSWATER2022 Page 10 of 16
NOTE:
5. Ruling Out Strength of reaction may be due to “dosage”
Look at the first completely negative cells ( panel cells that has 0 or If panel cells are homozygous, a strong reaction may be seen
no reaction) If panel cells are heterozygous, reaction may be weak or even
Go blood group by blood group or paired antigen non-reactive
Panel cells that are heterozygous should not be crossed out
Example case: because antibody may be too weak to react

6. Look what IS there (Look at the usual Ab reactivity)

Cell # 3 and 4 shows double dosage (homozygous Lea) Lea with


(+) reaction should be eliminated since the IS results should be
negative. Leb cannot be rule out since it has the same reaction
with IS results.
Cell # 5 and 7 shows single dose (heterozygous) cannot be ruled
out.
Cell #9 and 10 shows double dosage (homozygous) Leb with (+)
reaction can be ruled out.

7. Look for Matching Pattern


Single antibodies usually shows a pattern that matches one of the
antigens.
Multiple antibodies are more difficult to match because they often
show mixed reaction strengths.

#MLSWATER2022 Page 11 of 16
RULE OF THREE: FOR EXAMPLE:
Patient Serum must be: Let’s say you ran a panel and identified 3 different antibodies:
o Positive with 3 cells with the antigen anti-S, anti-Jka, and anti-P1
o Negative with 3 cells without the antigen o Selected cells could help…
o If there are not enough cells in the panel to fulfill the rule,
then additional cells from another panel could be used. Selected cells S Jka P1 IS LISS AHG
37
8. Use Special Techniques if Necessary #1 + 0 0 0 0 2+
#5 0 + 0 0 0 3+
9. Ensure statistical significance #8 0 0 + 0 0 0 (check cell +)

SPECIAL TECHNIQUES These results show that instead of 3 antibodies, there are actually
2: anti-S and anti-Jka
PHENOTYPING
In addition to the rule of three, antigen typing the patient red cells
can also confirm an antibody
NEUTRALIZATION/ INHIBITION
Only perform this if the patient has NOT been recently transfused
Other substances in the body and in nature have antigenic
(donor cells could react)
structures similar to RBC Antigens which can neutralize antibodies
If reagent antisera (of the suspected antibody) is added to the
present in serum, allowing separation
patient RBCs, a negative reaction should result.

NOTE: Individuals DO NOT make allo-antibodies against antigens SUBSTANCE ANTIBODY NEUTRALIZED
they have – Landsteiner Rule Anti P1
Hydatid cyst fluid, pigeon droppings,
turtledove’s egg whites
MULTIPLE ANTIBODIES
Selected cells are chosen from other panel or screening cells to Plasma/serum;secretor saliva Anti-Le
confirm or eliminate the antibody
Plasma or serum from Ch/Rg(+) individuals Anti-Chido; Anti-Rodgers
The cells are “selected” from other panels because of their
characteristics Anti-Sda
Urine, guinea pig urine
The number of selected cells needed depends on how may
antibodies are identified Human breast milk Anti0I

Secretor saliva ABH antibodies

#MLSWATER2022 Page 12 of 16
ENZYMES (PROTEOLYTIC) AUTOANTIBODIES
Can be used to enhance or destroy certain blood group antigens; Autoantibodies can be cold or warm reacting
Enzymes remove the sialic acid from the RBC membrane, thus A positive autocontrol or DAT may indicate that an auto-antibody is
“destroying” it and allowing other antigens to be “enhanced” present
Several enzymes exist: Sometimes the autocontrol may be positive, but the antibody
o Ficin (figs) screening may be negative, meaning something is coating the RBC
o Bromelin (pineapple)
o Papain (papaya) WHAT CAN THE DAT TELL US?
Although not always performed in routine pretransfusion testing,
DESTROYED ENHANCED NO EFFECT a positive DAT can offer valuable information
If the patient has been transfused, the patient may have an
MN, Duffy, Xg, JMH, alloantibody coating the transfused cells
ABO, Rh, Lewis, P, Ii,
Ss, Chido/Rodgers, Pr, Kell, Diego, Colton ,U If the patient has NOT been transfused, the patient may have an
Kidd
Lutheran autoantibody coating their own cells

One-stage: Enzyme is added directly to the serum/cell mixture Note: Auto-antibodies can sometimes “mask” clinically significant allo-
Two-stage: Panel cells are pre-treated with enzyme, incubated and antibodies, so it’s important to differentiate between auto- and allo-
washed. Patient serum is added to panel cells and tested antibodies

SULFHYDRYL REAGENTS
Cleave the disulfide bonds of IgM molecules and help differentiate COLD AUTOANTIBODIES
between IgM and IgG antibodies React at room temperature with most (if not all) of the panel cells
Good to use when you have both IgG and IgM antibodies and give a positive autocontrol
(warm/cold) The DAT is usually positive with anti-C3 AHG (detects complement)
Dithiothreitol (DTT) is a thiol and will denature Kell antigens Could be due to Mycoplasma pneumoniae, infectious mono, or cold
2-mercaptoethanol (2-ME) agglutinin disease
A combination of proteolytic enzymes and DTT
Denatures Kell, M, N, S, Duffy and other less frequent blood group
antigens
Does not denature the Kx antigen
Good for adsorption techniques
“frees” autoantibody off patient’s cell, so that autoantibody can
then be adsorbed onto another RBC

#MLSWATER2022 Page 13 of 16
AVOIDING REACTIVITY ELUTION (WHENEVER DAT IS POSITIVE)
Cold autoantibodies can be a nuisance at times. Here are a few ways to Elution techniques “free” antibodies from the sensitized red cells so
avoid a reaction: that the antibodies can be identified.
Use anti-IgG AHG instead of polyspecific. Most cold antibodies Testing the eluate is useful in investigations of positive DATs
react with polyspecific AHG and anti-C AHG because they fix o HDN
complement o Transfusion reactions
Skipping the IS phase avoids the attachment of cold o Autoimmune disease
autoantibodies to the red cells The red cells can also be used after elution for RBC phenotyping if
Use 22% BSA instead of LISS needed
If the antibodies remain, then prewarmed techniques can be When tested with panel cells, the eluate usually remains reactive
performed: with all cells if a warm autoantibody is present
Red cells, serum, and saline are incubated at 37° before being Types of Elution:
combined Acid elutions (glycine acid)
Autoadsorption is another technique in which the autoantibody is o Most common
removed from the patients serum using their own red cells o Lowers pH, causing antibody to dissociate
The serum can be used to identify any underlying alloantibodies Organic solvents (ether, chloroform)
o Dissolve bilipid layer of RBC
Heat (conformational change)
Freeze-Thaw (lyses cells)
WARM AUTOANTIBODIES
More common that cold autoantibodies
Positive DAT due to IgG antibodies coating the red cell
Again, the majority of panel or screening cells will be positive ADSORPTION
The Rh system (e antigen) seems to be the main target although Adsorption procedures can be used to investigate underlying
others occur alloantibodies
Cause warm autoimmune hemolytic anemia (WAIHA) ZZAP or chloroquine diphosphate can be used to dissociate IgG
Sources of Warm autoantibodies: antibodies from the RBC (may take several repeats)
o Idiopathic After the patient RBCs are incubated, the adsorbed serum is tested
o Known disorder (SLE, RA, leukemias, UC, pregnancy, with panel cells to ID the alloantibody (if present)
infectious diseases, etc) Two Types:
o Medications Autoadsorption
o No recent transfusion
o Autoantibodies are removed using patient RBCs, so
alloantibodies can be identified
Allogenic (Differential) adsorption
o If recently transfused
o Uses other cells with the patients serum

#MLSWATER2022 Page 14 of 16
ANTIBODY PANEL VS. ANTIBODY SCREEN
CHLOROQUINE DIPHOSPHATE o Group O reagents but with antibody panel it uses 10-15 or 8-20 vials
Quinilone derivative often used as an antimalarial or donors blood compared into 2-3 vials in antibody screening.
May not remove autoantibody completely from DAT positive cells o Patient serum is also used just like in antibody screening.
Partial removal may be enough to antigen type the cells or to be o It also involves 3 phases: immediate spin, 37° Celsius, and
used for autoadsorption of warm autoantibodies antihuman globulin
o We use 10 panel cells, the number 11 is usually the auto control
o Group O cells are used so that anti-A and anti-B will not interfere in
the detection of antibody to the other blood group system
ADD-ONS FROM MA’AM JOANA’S DISCUSSION o Each of the panel cells have been antigen type
When do we need to identify the antibody?
ANTIGRAM/WORKSHEET
COMMON REASONS TO IDENTIFY AN ANTIBODY PANEL TESTING o The table is called the antigram
o Uses Antibody Panels - are used to identify an unexpected antibody o antigram or worksheet is a listing of the antigen make up of each of
detected by the antibody screening the screening cells or panel cells that is provided with a lot of
o This is usually done following a positive antibody screening when screening cells. This can be used as a worksheet during antibody
testing would suggest a presence of new antibody and confirm a detection process
previously identified antibody o (+) positive or plus sign symbol: refers to presence of antigen
o (0) zero: refers to the absence of antigen
o Auto controls are located below, it is used to determine the
Why do we need to identify the antibody present in a reaction?
antibody present is an autoantibody or alloantibody
o it is needed for transfusion processes
o it is essential component for the compatibility testing  There is the presence of autoantibody if the auto control with
o identification of any unexpected antibodies in the patient’s serum the patient’s RBC will result a (+) positive reaction
 If (0) zero or no agglutination that would occur during the auto
In antibody panel testing, it is basic to know what is alloantibody and control testing would mean that the antibody present is
autoantibody alloantibody
o Alloantibodies – are antibodies against RBC agent not present on o It is also needed to check in the antigram the reaction in different
patient’s own RBC phases: immediate spin, 37° Celsius, and antihuman globulin
o Autoantibodies- are antibodies against the RBC antigen present on o 10 panel: a drop in each panel cells and 2 drops of patient’s serum
patient’s own RBC
IMMEDIATE SPIN PHASE
RBC screening o Detect clinically insignificant cold antibodies usually IgM in nature
o uses 2-3 screening cells to detect if any antibodies are present in the and it involves immediate centrifugation of the mixture at room
serum temperature for 20 seconds using the centrifuge
o it follows the concept about when detecting/identifying the  There are some laboratories that will not require immediate
antibodies, test the patient’s serum (contains the unknown), weigh spin phase they directly undergo LISS phase and anti-human
the reagents RBC (contains the known) globulin

#MLSWATER2022 Page 15 of 16
37° CELSIUS WITH ENHANCEMENT MEDIUM DIFFERENT GRADING FOR AGGLUTINATION
o it detects the warm reactive IgG antibodies 1 CLUMP 4+
 Enhancement Media are potentiators that added to the cells IF DISTANCED 3+
serum mixture before the 37° Celsius or incubation to increase SCATTERED 2+
the sensitivity of the test system HAS AGGLUTINATION BUT VERY 1+
MINIMAL
DIFFERENT ENHANCEMENT MEDIA NO AGGLUTINATION REPORTED AS NEGATIVE
1. 20% Albumin
DO NOT FORGET TO ADD THE CHECK CELLS TO ANY NEGATIVE AHG
o Incubation period 30-60 minutes, works in decreasing zeta
o Example: Encounter a reaction that can be only seen in Immediate
potential Spin Phase
o Low Ionic Strength Solution/ Saline Solution: Incubation time 10- o No reaction was recorded for the LISS and no reaction was also seen in
15 minutes and it contains .2% of Sodium Chloride. Meaning it anti-human globulin.
has low ionic content compared to the normal saline solution o To confirm that the procedure is performed correctly, it doesn’t need
that contains glycine, dextrose or glucose in addition to saline to repeat and check cells must be performed. Since check cells are added
reduce the zeta potential in all negative tubes
o increases the amount of the antibody taken up by the red cell  Check cells – are cells coated with IgG and should react
during the sensitization process positively with anti-human globulin reagent within the tube.
o Sensitization – is the first page in the agglutination process If the check cells are negative, procedure was not
performed correctly and should be repeated
2. Polyethylene Glycol
-END OF TRANSCRIPTION -
o Works in removing water from the test system thereby

concentrating any antibodies that are present in the reaction.
More sensitive compared to LISS and 22% albumin
 Disadvantage: don’t usually used in high motile patients
having high protein levels just like in the cases of multiple
myeloma since it can interfere in the testing procedure

3. Antiglobulin phase
o Detects non-agglutinating warm reactive antibodies that are
possibly not detected with 37° Celsius incubation period.
o It would also detect antibodies that has been coated or
sensitized, RBC that did not show any visible agglutination.

#MLSWATER2022 Page 16 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 1)
MLS 18, April 6, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
Has regulatory authority over drugs, biologics and devices as
Legend: mandated by the Federal Food, Drug and Cosmetic (FDC) Act of
Transcription Bullet 1938 and the Public Health Services (PHS) Act of 1944.
Notes/Module Packet Bullet Blood establishments in the US are therefore:
 PPT • Regulated by the FDA
• Required to follow federal regulations provided by the FDA in
MODULE OUTLINE
the composite form of the CFR (Code of Federal Regulations)
I. Organizations that Regulate or Accredit the Immunohematology
• Inspected at regular intervals by agents of the FDA
Laboratory
Blood establishments that ship products across state lines must be
II. Blood Donor Selection
licensed by the FDA, while blood banks and hospital transfusion
III. Types of Donation
services that operate within state boundaries must be registered
IV. Blood Component
with the FDA
V. Safe Storage of Blood
Hospital transfusion services that do not perform any
VI. Packing Blood Components for Transportation
manufacturing processes as defined by the FDA are not required to
be licensed or registered, though they must be in compliance with
LEARNING OUTCOMES applicable portions of the CFR and state regulations.
At the end of this module, the student shall be able to:
1. Identify the organizations that regulate or accredit the A government agency that regulates and establishes blood products
immunohematology laboratory. and its guidelines, transportation temperature, storage temperature
2. State the minimum acceptable levels for the following tests in and the products used in blood bank laboratory
allogenic & autologous donation Established CBER (Center for Biologics Evaluation and Research) is
3. Identify the medical history information that would cause responsible for the regulation of collection of the blood and blood
permanent deferral or temporary deferral and state the length of components that is being used in the transfusion and for the
deferral period. manufacture of the pharmaceutical derived from the blood and
4. Explain the reasons for permanent or temporary deferral given the blood components. It developed and enforces quality standard and
various medical conditions. inspection in blood establishments and monitor reports about
errors, accidents and adverse clinical events that occurs in the blood
transfusion practice. In hospital transfusion services it will not
ORGANIZATIONS THAT REGULATE OR ACCREDIT THE
perform any manufacturing processes defined by the FDA are not
IMMUNOHEMATOLOGY LABORATORY
required to licensed or registered.
FDA
Food and Drug Administration AABB (American Association of Blood Banks)
An organizational arm of the Department of Health and Human A professional organization which serves a number of key functions
Services (HHS) often referred to as the Agency. for the blood industry, including education, supporting science and
research and its dissemination, and public advocacy.

#MLSWATER2022 Page 1 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
• The National Blood Foundation (NBF), a subsidiary of AABB,
was created with the sole mission of providing seed funding Donor Requirements
and financial support for scientific inquiry related to 1. Identification card
transfusion medicine Includes photographic identification of the donor

College of American Pathologists (CAP) 2. Donor registration information


Participates in laboratory accreditation and proficiency testing. Includes full name, date and time of donation, address,
The CAP program uses a system of peer review, and participating telephone/cell phone number, sex, age and date of birth
institutions must provide inspectors for future accreditation visits.
3. Consent Form
BLOOD DONOR SELECTIO N
Two questions that must be answered before proceeding with the 4. Additional Information
blood donation: The name of the patient whom the blood is intended
1. Will a donation of approximately 450 mL of whole blood at this time Race of the donor for the rare phenotype
be harmful to the donor? CMV status
2. Could blood drawn from this time potentially transmit a disease to
the recipient Calculations for the Blood Volume:
 Calculate adjusted blood volume and anticoagulant:
If “yes”: Defer donor: temporarily or permanently In athletes, pulse rate of <50 bpm is considered normal and not a factor
for deferral
If “No”: Proceed to Blood Donation
Volume to Collect:
General Requirements for Donation
𝑑𝑜𝑛𝑜𝑟 ′ 𝑠 𝑤𝑒𝑖𝑔ℎ𝑡 𝑖𝑛 𝑘𝑔
= 𝑥 450 𝑚𝐿
General Appearance: Observe the prospective donor for the 50 𝑘𝑔
presence of excessive anxiety, drug or alcohol
influence, or nervousness.
Age At least 17-18 years old Reduced Volume of Anticoagulant:
Oral Temperature < 37.5 °C or 99.5 °F
Blood Pressure Less than or equal 180/100 mmHg 𝑉𝑜𝑙𝑢𝑚𝑒 𝑡𝑜 𝐶𝑜𝑙𝑙𝑒𝑐𝑡
= 𝑥 63 𝑚𝐿
450 𝑘𝑔
Hemoglobin ≥12.5%
Hematocrit ≥ 38%
Pulse 50-100beats/minute Volume of Solution to be removed:
Weight ≥ 110 lbs or ≥ 50 kg
= 63 𝑚𝐿 − 𝑟𝑒𝑑𝑢𝑐𝑒𝑑 𝑎𝑚𝑜𝑢𝑛𝑡 𝑜𝑓 𝐴𝑛𝑡𝑖𝑐𝑜𝑎𝑔𝑢𝑙𝑎𝑛𝑡
Volume Maximum of 525 mL can be collected
WB Donation Maximum of 10.5 mL of blood /kg of donor See Appendices for Sample Problem
weight

#MLSWATER2022 Page 2 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Blood Collection Process Position needle at 10-20° angle
1. Donor Registration 16-gauge needle usually used for blood donation; instruct
donor to open-and-close their fists every 10-20secs
2. Educational material is distributed to the donor. Mix the unit periodically 1-2 times per minute.
 If the prospective donor shows symptoms of an infectious disease,
o Blood Mixer – automatically mixes the blood with
the donor is excluded from the donation.
anticoagulant
Explain to the donor what might happen to his blood since it will
o If there is no blood mixer, the medical technologist will do
undergo different blood test
the mixing manually.
They must also sign the consent form allowing for the blood
Amount of blood collected: 450mL ± 10%
collection as well as other blood tests
Duration of collection: 7-10 minutes
Medical technologists must further explain to the donor the
o If the correct duration time is not observed, the blood
other blood tests to be conducted to his blood
collected would not be suitable for platelet
concentration/concentrate preparation, FFP, or
3. Actual Donor selection/ Identification cryoprecipitate (>15mins of blood collection would make
No fasting is required for blood donation, but donor must eat the blood not viable, especially in cryoprecipitate).
Donors who have not eaten food for 4hrs or more are required
to eat before blood collection Color Coding for Blood Bags

4. Blood Collection FDA (1985) *RA 1517


 Use Aseptic Technique: Blood Type Color Label Blood Type Color Label
o PVP Iodine compound or chlorhexidine gluconate & A Yellow A Blue
Isopropyl alcohol B Pink B Yellow
o Scrub site at least 4 cm in all directions for 20 seconds AB White AB Pink
o Apply tourniquet or blood pressure cuffs: 40-60 mmHg O Blue O White
o Position the needle at 10-20 ° angle *Blood Bank Law; the standard/ what used
 Mix the unit periodically 1-2 times per minute
 Amount of blood collected: 450 mL ± 10% Donor Deferral
 Duration of collection: 7-10 mins
o Component must be prepared after collection TEMPORARY DEFERRAL
 EXAMPLE: Donor has received a blood transfusion; defer for 12
Blood collection facilities confirms the identity of the donor months from date of transfusion
through the photographic identification such as the use of  Tooth Extraction
showing their driver’s license for confirmation  Persons who had experienced convulsions
The antecubital fossa should be free from lesions, scars and
track marks INDEFINITE DEFERRAL
During blood collection 2 blood bank personnel, 1 Medical  These donors may be eligible to donate autologous blood
Technologist and 1 Head of Blood Bank is required to be The donor is unable to donate their blood for some reason and
present. They do interview and look for multiple needle marks, there is no specified period of time due to regulatory requirements;
unexplained weight loss or if the patient is immunosuppressed, but donors are still eligible to donate autologous blood
oral thrush or Kaposi sarcoma
#MLSWATER2022 Page 3 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
PERMANENT DEFERRAL
TEMPORARY DEFERRAL
 EXAMPLE: Donor states that he or she has hepatitis C; defer Deferral if the donor received the following vaccine:
permanently 2-week deferral Smallpox, Polio, Measles, Mumps, Influenza
The donor will never be eligible to donate blood for someone else; 4-week deferral German Measles, Chicken-pox
donor is only eligible to donate autologous blood
12-month deferral Rabies vaccine if given after the bite of a
o Definite disease or habits strongly associated with bloodborne
rabid animal
pathogens
May donate if afebrile Diphtheria, Pertussis, Typhoid, Tetanus,
o Narcotic addiction
Cholera, Influenza
o Alcohol addiction
1 Year  Skin penetration w/ instruments
o Received pituitary growth hormones of human origin
(Possible exposure to contaminated w/blood (surgery)
o History of chaga’s disease, malaria, or babesiosis; cancer,
Hepatitis, HIV or Malaria)  Closed contact with persons with viral
leukemia, lymphoma
hepatitis
o Persons w severe thrombocytopenia
 (+) STS- 12 months from completion of
therapy
PERMANENT DEFERRAL  Traveled to endemic area for malaria w/
Definite disease or Viral Disease or w/o antimalarial drugs
habits strongly  Hepatitis after the age of 11
 ≥72 hours in correctional institution
associated with  (+) confirmatory test for HbsAg
bloodborne pathogens  Repeatedly (+) test for anti-Hbc  Female donor who had sex with bisexual
 Gave only unit to recipient who men
developed post transfusion hepatitis and  Anyone who had given someone money
HIV or drugs in payment for sexual contact
 Present and Past infection with Hepa C 3 Years  Visit/ immigrant from area endemic for
and HIV (Possible exposure to malaria
 Narcotic addiction malaria)  Had malaria, but presently asymptomatic
 Alcohol addiction
Other diseases: **plasma preparation w/o red cells are
 Received pituitary growth hormone of exempted from these restriction
human origin No deferral if afebrile  Diphtheria, pertussis, typhoid, tetanus,
 History of Chaga’s disease, babesiosis or cholera, influenza, previous history of tb
malaria that has been successfully treated and is
 History of cancer, leukemia, or lymphoma no longer active
 Person’s with severe thrombocytopenia

#MLSWATER2022 Page 4 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Intake of Medication injections given over
a 6-month period
Medication Indication Deferral Period Unlicensed Vaccine Research Proposal 12 months unless
Proscar (Finasteride) Prostate Gland 2 months otherwise
Avodart (Dutasteride) enlargement 6 months indicated by
Propecia Baldness 2 months medical director
Accutane (Amnesteem, Severe Acne 2 months
Claravis, Sotret, How it affects donation?
Isotretinoin)  HBIG does not prevent Hepatitis B in every case
Soriatane (Acitretin) Severe Psoriasis 3 years
Tegison (Etretinate) Permanent Contraceptive Pills
 May donate anytime
How it affects donation?
 These medications can cause birth defects. Donated blood with high Highly Allergenic Drugs like Penicillin, Aspirin, and others
enough levels to damage the unborn baby if transfused to a  May donate anytime provided the blood collected will not be used
pregnant woman. to prepare platelets
 Once medications are cleared from the body, one can donate again  Donor may donate only after 24 hours of taking the medications.

Medication Indication Deferral Period Piroxicam


Growth Hormone from Delayed impaired Permanent  Deferred until TB is completely cured
Human Pituitary Glands growth (used only until
1985) TYPES OF DONATION
Insulin from cows Indefinite
ALLOGENIC DONATION
(Bovine or beef insulin)
Genetically different individual but same specie
Blood is taken from an individual of the same specie as the recipient
How it affects donation?
 Can lead to development of rare nervous system disorder called CJD
(Creutzfeldt-Jakob Disease) REQUIREMENTS FOR ALLOGENIC DONATION
 If Insulin was imported from countries infected in which Mad Cow
Disease had been found, it could contain material from infected Philippine Standard AABB Standards
cattle Age 16-65 years old At least 17 years old
 MCD can be transmitted through blood transfusion Temperature ≤37.5 °C or ≤99.5 °F ≤37.5 °C or ≤99.5 °F
Pulse Rate 60-100 bpm 50-100 bpm
Medication Indication Deferral Period Hemoglobin ≥ 12.5 g/dL ≥ 12.5 g/dL
HBIg  Following exposure 12 months after
Blood Pressure
to hepatitis B receipt of vaccine
 Systolic 90- 160 mmHg ≤ 180mm Hg
 Different for
 Diastolic 60-100 mmHg ≤ 100 mm Hg
Hepatitis B vaccine,
which is a series of 3

#MLSWATER2022 Page 5 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
AUTOLOGOUS DONATION 3. INTRAOPERATIVE COLLECTION
Autologous means “self”  Involved collecting shed blood from the surgical site, a device that
Blood is given to the recipient came from the recipient himself utilizes vacuum is used to collect shed blood.
REQUIREMENTS FOR AUTOLOGOUS DONATION  Blood is the washed with saline and concentrated to reach
hematocrit of 50-60%, then reinfusing those cells immediately.
Collection and Reinfusion of lost blood during a surgery
Age No age requirement
Hemoglobin 11 g/dL
Hematocrit 33% 4. POSTOPERATIVE COLLECTION
 General Condition: Patient should have no condition predisposing to  Collected from a drainage tube placed at the surgical site.
bacteremia or any form of severe cardiovascular/ pulmonary  It is reinfused with or without processing, via a microaggregate filter
condition. to screen out any debris
 Single unit is removed at a time, with at least 3 days intervals
 Final phlebotomy must be at least 72hrs before surgery DIRECT DONATION
 A direct donation is a unit collected under the same requirements as
those for allogenic donors, except that the unit collected is directed
TYPES OF AUTOLOGOUS DONATION towards a specific patient.
The patient selects his/her own donor for an anticipated non-
1. PREOPERATIVE COLLECTION emergency transfusion
 Removal of blood 5-6 weeks immediately preceding a scheduled, Donor is typically a friend or relative of the patient
elective surgical procedure
The storage of blood and its components before an elective surgery; APHERESIS DONATION
blood is used during or after surgery  Aphaeresis= “taking away”
o Possible Problems: presurgical anemia or hypovolemia can  An effective mechanism for collecting a specific blood component
occur, clinical identification errors, outdating of liquid-stored while returning the remaining whole blood components back to the
blood, homologous blood transfusion instead of autologous patient
 Amount of time for a particular procedure can range from 45-120
2. ACUTE NORMOVOLEMIC HEMODILUTION minutes
 Collection of whole blood with the concurrent infusion of crystalloid  ACD: Most common and Primary Anticoagulant in Apheresis
or colloid solutions procedure
 The idea is that the patient bleeds more dilute blood during the It is the withdrawal of blood from a donor removing selected
procedure, and the patient’s heart may pump more effectively due components and reinfusion of the remaining components back to
to decreased blood viscosity the donor
Isovolemic Hemodilution
o Multiple units of blood are collected; the blood is reinfused Methods of Centrifugation
near-end the surgery
1. INTERMITTENT FLOW CENTRIFUGATION
 The last unit collected must be the first unit to go back
 Blood is drawn and reinfused through the same needle
 Once the desired component is separated, the remaining
components are reinfused to the donor

#MLSWATER2022 Page 6 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
The blood is processed in batches or cycle and requires only 1
venipuncture.  Donor’s RBCs should be ABO- and Rh-compatible, relatively fresh
Spins, process in the machine and gives back the unused part to the (leukoreduced, negative for hemoglobin S and partially phenotype-
donor. The blood is return to the same site where it was extracted. matched for the Rh (C, c, E, e) and K1 antigens
To stop the blood from coagulating, anticoagulant is automatically
mixed with the blood as it pumps the blood into the apheresis WBC PHERESIS (LEUKAPHERESIS)
machine. The granulocyte concentrate from leukapheresis are needed for
patients who have conditions such as severe neutropenia or
2. CONTINUOUS FLOW CENTRIFUGATION (CFC) conditions that are unresponsive to antibiotic.
 Involve withdrawal and processing and reinfusing of blood to the Storage time is 24 hours at 20-24°Celsius.
individual simultaneously Compatibility testing is typically performed for WBC pheresis.
PLASMAPHERESIS (PLASMA EXCHANGE)
 Equivalent of at least two whole-blood derived plasma units
o Infrequent plasmapheresis
o Frequent or Serial
 FDA recommends 12 L (14.4 L for donors weighing more than 175
pounds) as the maximum allowable plasma volume donated per
year
Maximum of 2 donation in a 7 day period with 2 days gap period is
allowed.

THERAPEUTIC PROCEDURES
 The rationale of Therapeutic Apheresis (TA) is based on the
following:
o A pathologic substance exists in the blood that contributes
Types of Apheresis to a disease process or its symptoms
PLATELETPHERESIS (THROMBOCYTOAPHERESIS) o The substance can be more effectively removed by
 Donor’s platelet count: at least 150,000/ µL apheresis than by the body’s own homeostatic mechanisms
 Donor should have not taken aspirin 3 days before donation
 Interval of at least 2 days 1. Therapeutic Plasma Exchange (TPE)
 pH ≥ 6.2  Removal and retention of the plasma, with return of all cellular
components to the patient.
 To remove the agent in the plasma, such as an antibody, toxin, or
RBC PHERESIS (ERYTHROCYTAPHERESIS) abnormal protein, that is causing the clinical symptoms.

REQUIREMENTS FOR ERYTHROCYTAPHERESIS Factors Removed by Therapeutic Plasmapheresis


Height Weight Hct Immune Complexes SLE
Male 5’11” 130 lbs 40% Alloantibodies Antibody
Female 5’5” 150 lbs 40% Autoantibody Gullain-Barré syndrome
#MLSWATER2022 Page 7 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Immunoglobulin causing Waldenstrōm’s macroglobulinemia NAUSEA OR VOMITING
hypersensitivity  Management:
Protein-bound toxins or drugs Amanita mushroom poisoning, 1. Instruct the donor to breathe slowly
barbiturate poisoning 2. Apply cold compresses to the forehead
LPPs Familial hypercholesterolemia, 3. Turn the donor’s head to one side and provide an appropriate
hypertriglyceridemia receptacle
Phytanic Acid Refsum’s disease 4. The donor may be given water after vomiting has ceased

LOSS OF CONSCIOUSNESS
2. Therapeutic Platelet Pheresis  Management:
 Thrombocytosis (at least 500,000/ µL) can occur in myeloproliferative 1. Check vital signs frequently
disorders (ET, PV, CML) or as a reactive process in response to 2. Administer 95% O2 and 5% CO2
splenectomy, infection, chronic inflammation, or malignancy
SEVERE REACTIONS
3. Therapeutic Leukapheresis
CONVULSIONS
 Elevated level of WBCs placed the patient at risk for complication
 Management:
associated with leukostasis, including organ dysfunction due to the
1. Call for help immediately; notify blood bank physician
formation of microthrombi in the pulmonary and cerebral
2. Try and restrain the donor to prevent injury to self or others
microvasculature
3. Ensure an adequate airway

STEM CELL PHERESIS


CARDIAC OR RESPIRATORY
 Management:
DONOR REACTIONS
1. Perform CPR until medical help arrives
MILD REACTION
HEMATOMA
SYNCOPE  Management:
 Management:
1. Remove the tourniquet and withdraw needle
2. Place cold compression on the donor’s forehead POST DONOR CARE
3. Raise the donor’s legs above the level of the head  Raise arm and apply pressure on the puncture site after collection.
4. Loosen tight clothing and secure airway  Rest after blood collection, reclining for few minutes, then sit upright
5. Monitor vital signs and allow to talk.
 Instruct the donor to drink a lot of water, refrain from smoking and
TWITCHING OR MUSCLE SPASMS avoid strenuous work or driving.
 Management:
1. Disengage the hyperventilation sequence by conversing with
the donor and having the donor breathe into a paper bag.

#MLSWATER2022 Page 8 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
NOTES FROM MODULE PACKET:
Blood Donor Selection Key principles of blood donor selection:
The primary responsibility of a blood transfusion service is to The health and safety of the donor as well as the recipient must
provide a safe, sufficient and timely supply of blood and blood be safeguarded
products Only individuals in good health should be accepted as donors of
The purpose of blood donor selection is to: whole blood and blood components
o Protect donor health and safety by collecting blood only The selection of blood donors should be based on regularly
from healthy individuals reviewed selection criteria, without discrimination of any kind
o Ensure patient safety by collecting blood only from donors including gender, race, nationality or religion
whose donations, when transfused, will be safe for the A prospective donor’s health status and medical history should be
recipients evaluated for each donation, on the day of donation prior to
o Identify any factors that might make an individual blood collection
unsuitable as a donor, either temporarily or permanently The blood donor selection (BTS) should provide appropriate
o Reduce the unnecessary deferral of safe and healthy donors donor information and a simple donor questionnaire for health
o Ensure the quality of blood products derived from whole and risk assessment and obtain the donor’s informed consent to
blood and apheresis donations blood donation
o Minimize the wastage of resources resulting from the Staff should be suitably qualified and trained in the donor
collection of unsuitable donations. selection process
Blood donors have a responsibility to self-defer if they are aware of Good communication should be established between the BTS
having been exposed to any risk of an infection or a known health staff and the donor, and donor confidentiality should be assured
condition or treatment that could influence their suitability to The BTS has a duty of care to provide counselling to all deferred
donate blood. donors and referral for their further management.
Blood donors also have the right to withdraw at any stage of the
donation process. Steps Involved in The Donor Selection Process
A donor questionnaire is the key tool in donor selection for Compliance with all donor selection criteria is crucial to ensure a
assessing donor health and safety and for reducing the risk of safe blood donation process and outcomes.
transmission of infection, in particular for infections for which no
Donor registration
suitable screening tests are available.
All prospective donors who meet the general criteria for blood
o The use of a donor questionnaire prompts donor selection
donation such as age and good health should be registered when
staff to ask important questions and carefully assess the
they attend a blood donation session, even if they are subsequently
donor’s health.
not accepted for donation.
o By presenting all relevant information in a standard format,
Essential donor registration information includes the individual’s full
a donor questionnaire facilitates decisions on the
name, date of birth, gender and contact details.
acceptance or deferral of the donor.
During donor registration, prospective donors should be provided
Effective public information and donor education are the first steps
with donor information and education materials and the donor
in the process of donor selection.
questionnaire, which should be completed on each occasion of
donation.

#MLSWATER2022 Page 9 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Pre-donation information donor selection process; and donor’s duties, responsibilities and
The process of donor selection begins even before donors come to rights
give blood through public awareness campaigns and donor Options for the donor to decide about blood donation prior to
education. proceeding further, to withdraw or self-defer at any time during
At the donation session, pre-donation information should be or after the donation process, without any undue embarrassment
provided either orally or through printed, graphic, audio-visual or or questioning
online materials, presented in a simple and clear format and in Transfusion-transmissible infections, including HIV, HBV, HCV and
appropriate languages. syphilis, routes of their transmission, natural history and
Pre-donation information provides an opportunity for the prevention; types of screening tests performed; and window
prospective donors to know about health conditions or high-risk period of infection and alternative testing sites for individuals
behaviour that would make them unsuitable to donate blood. seeking to ascertain their infection status
This information assists the donors in deciding whether to self- Possible consequences for donors and the donated blood in the
defer; case of abnormal TTI test results; the mechanism for notification
• It may also assist in donor return if they understand the about abnormal test results and post-donation counselling,
reason why they should not donate blood on this occasion assurance of confidentiality and if necessary, referral for further
testing, treatment and care
Objectives of Pre-donation Information: The possibility of adverse donor reactions.
Increase donor awareness of the donor selection criteria, the
process of blood donation and the tests that will be performed on Completion of donor questionnaire
donors’ blood Each prospective blood donor should complete a donor
Encourage prospective donors to inform the BTS of any medical questionnaire to provide information in relation to the donor
conditions or TTI-related risks that may affect their suitability to selection criteria defined in the national guidelines.
donate blood In most situations, the donor questionnaire is given to donors at the
Encourage individuals to self-defer from blood donation if they time of registration for completion before the donor interview and
recognize that they are not suitable to donate blood due to assessment.
general health or medical conditions or risk for TTI. Alternatively, the donor questionnaire may be sent to the donor’s
residence to be completed before donation.
Pre-donation Information Coverage: • This has the advantage of allowing donors time to think about
Nature and use of blood and its components; the need for the answers and saves time at a blood donation session.
voluntary nonremunerated blood donors; and the importance of • However, donors may misunderstand some of the questions
maintaining healthy lifestyles and self-defer for the wrong reasons.
The blood donation process, including the donor questionnaire, The donor questionnaire may also be administered electronically as
donor medical history, health and risk assessment, venipuncture, a computer-based questionnaire.
blood collection as whole blood or apheresis procedure, post- It is essential that donors are aware of the importance of the
donation care and the screening tests performed on donated questionnaire, the significance of the questions and the need for
blood providing accurate information
Rationale for the donor questionnaire and pre-donation health The information provided by the donor can then be further
assessment and the importance of donor compliance in the elaborated on during the interview.

#MLSWATER2022 Page 10 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Donor interview and pre-donation counselling The basic health check also enables an assessment to be made of
The completed donor questionnaire should be reviewed prior to any physical disabilities that may impede the donation process, such
donation in a one-to-one confidential interview between the donor as:
and a donor selection staff member so that an assessment can be • Mobility: the donor should be able to easily access the donor
made of the donor’s general health, medical history and any TTI bed or couch
risks. • Sight or hearing impairment: assistance should be provided
It also provides an opportunity to check whether the donor has by a staff member.
understood the questions and has answered them correctly. Issues that require special attention during donor health and risk
Assurance about the confidentiality of the donor’s medical history is assessment include:
essential. • The prevalent culture and context of the environment for
Pre-donation counselling is an integral part of the donor interview. donation; in some situations, a donor may simply be
It enables donor selection staff to: overawed by the medical setting and procedures
• Check that the donor has understood all questions and • The provision of sufficient privacy and assurance of
responded accurately to the questionnaire confidentiality to make the donor comfortable when
• Answer the donor’s questions and provide reassurance in case answering probing and sensitive questions
of anxiety • Identifying and overcoming language barriers or lack of
• Explain reasons for any deferral and give advice about further understanding of questions in the donor questionnaire
medical care, if needed • Ensuring good communication by using simple jargon-free
• Ensure that the donor is able to give informed consent to language and explaining any medical terms.
donate and recognizes that his/her signature is an affirmation
that responses provided to the questionnaire are accurate. Informed consent
Informed consent is a voluntary agreement given by the prospective
Donor health and risk assessment donor to the donation of blood, to the testing of a blood sample for
The assessment of donor suitability and deferral, where TTI, for the transfusion of the donated blood to patients and if
appropriate, aims to exclude donations from individuals at risk of required, for the use of the blood for additional tests, quality
TTI, particularly from those with recently acquired infection that assurance or research purposes.
cannot or may not be detected by routine screening tests or with To obtain informed consent, the BTS should provide the following
infections for which no effective blood screening tests are available. minimum information to the potential donor:
The donor assessment not only enables the review of the donor’s • The donation process and potential adverse donor reactions
medical history and medications, but also provides an opportunity • The tests that will be performed (TTI and others) on the
for a basic health check to assess whether the donor is in general samples taken from the donated blood and the reasons for
good health. these tests
Any signs of debility, under-nutrition, pallor, jaundice, cyanosis, • Confidentiality of all personal information, including test
dyspnea or intoxication from alcohol or drugs should also be noted results.
Physical examination, weighing and/or measurement of vital signs The donor should sign and provide informed consent to the
(pulse, blood pressure) are part of the basic health check and are donation of blood or blood components on a voluntary basis.
carried out at this stage. Informed consent signifies that the donor has understood the
The venipuncture site should be examined to check that the donor’s questionnaire, has provided accurate answers and is willing to
veins are accessible and suitable to enable easy venipuncture. donate blood.

#MLSWATER2022 Page 11 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
It also indicates that the donor understands the blood donation Criteria for Blood Donor Selection
process, the possibility of adverse reactions to blood donation, the
Age
risks of the transmission of infections through donated blood and
The lower age limit for blood donation in most countries is 18 years,
the implications of any abnormalities that may be detected during
although in some countries national legislation permits 16–17 year-
the donation process and blood screening, and is providing consent
olds to donate provided that they fulfil the physical and
for post-donation notification and counselling, if detected to have a
haematological criteria required and that appropriate consent is
positive viral infection marker or any other abnormality.
obtained.
• Adolescents of either gender are at risk of iron deficiency
Donor Deferral:
during the pubertal growth spurt when the average daily total
Donors who do not to meet the selection criteria should be
requirement of absorbed elemental iron is 1.50 mg/day for
deferred on a temporary or permanent basis.
males aged 15–17 years and 1.62 mg/day for females
All deferred donors should be treated with respect and care in a
Upper age limits for blood donation of between 60 and 70 years
confidential manner and should be given a clear explanation of
have been implemented in the past because of concerns regarding
the reason for deferral and an opportunity to ask questions.
the increasing incidence of cardiovascular disease with age and the
They should be informed whether the deferral is to safeguard
potential risk of adverse reactions, which are more likely in first-
their own health and/or that of the recipient.
time donors.
It is the responsibility of the BTS to ensure that donors who are
• The usual upper age limit for blood donation is 65 years
deferred due to medical conditions are referred for further
• First-time donors older than 60 years and regular donors over
investigations and management, as appropriate.
the age of 65 may be accepted at the discretion of the
Temporarily deferred donors should be advised on when they
responsible physician
could donate and encouraged to return
• First-time donors over 60 years should make their first
A donor deferral registry (DDR) is a confidential list of donors
donation at a donation site where a physician is available
who are positive for a transfusion-transmissible infection and
who have been permanently deferred.
A DDR is used to monitor the incidence and prevalence of such DONOR APPEARANCE AND INSPECTION
infections in the donor population and may also assist in Prospective donors should be accepted only if they appear to be in
identifying areas that require strengthening in the donor good health and comply with donor selection criteria
selection process. The prospective donor should appear generally well and should not
be febrile, breathless or suffering from a persistent cough.
The colour of exposed skin and mucous membranes should be
normal, with no jaundice, cyanosis, flushing or pallor, and no signs
of skin infection, rash or obviously enlarged lymph nodes.
• If body piercings or tattoos are present, the risk of
transfusion-transmissible infections (TTI) should be assessed

MINOR ILLNESSES
Individuals with a history of recent infection: defer for 14 days
following full recovery and cessation of any therapy, including
antibiotics

#MLSWATER2022 Page 12 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
• Minor non-specific symptoms (e.g. general malaise, pain, DONOR IRON STATUS
fever, headache, cough, diarrhoea) may indicate the presence Haemoglobin screening safeguards anaemic individuals from
of an acute infection that may be transmissible by transfusion. donating blood and also protects returning donors from donation-
• Donors should be asked to confirm that they are free from induced iron deficiency (DIID), the depletion of iron stores by
such symptoms on the day of donation and that they have repeated donations.
fully recovered from any recent infection(s). Collecting a unit of blood from a donor with a normal haemoglobin
• Individuals suffering from minor illnesses and not feeling well level also provides good quality blood components, with adequate
should not donate blood. and consistent haemoglobin content in the collected blood.
In determining the lower limits of haemoglobin for whole blood
WEIGHT donation and implementing haemoglobin screening, the BTS should
Prospective donors of whole blood donations should weigh at least consider:
45 kg to donate 350 ml ± 10% and 50 kg to donate 450 ml ± 10% • A haemoglobin level of not less than 12.0 g/dl for females and
Prospective donors of apheresis platelet or plasma donations should not less than 13.0 g/dl for males as the threshold
weigh at least 50 kg Only sterile disposable lancets should be used for blood sampling
Prospective donors of double red cell apheresis donations should
have an estimated blood volume of more than 5 litres; this FLUID INTAKE AND FOOD
requirement is generally met by non-obese individuals weighing Most BTS guidelines recommend that donors should maintain their
more than 70 kg. usual food and fluid intake before donation but should avoid heavy
or fatty meals which may result in a lipaemic donation that may
VITAL SIGNS need to be discarded
PULSE The BTS should consider providing 500 ml drinking water to donors
• A normal pulse rate of 50–100 per minute and a regular before donation to minimize the risk of vasovagal reactions
rhythm are indicators of good health; many BTS recommend
that these are examined prior to donation. PREGNANCY, LACTATION AND MENSTRUATION
• The ability to detect significant abnormalities of pulse rate or The average woman needs about 350–500 mg additional iron to
rhythm depends on the skill and experience of staff. maintain iron balance during pregnancy.
BODY TEMPERATURE Female donors should be deferred during pregnancy and for a
• A prospective donor who is febrile – defined as a core oral sufficient time after delivery (or following abortion or miscarriage)
temperature more than 37.5°C– is by definition unwell and and during lactation to allow for the recovery of iron stores.
should be deferred. Menstruation is not a reason for deferral. However, women who
• Fever can indicate any number of medical conditions and report regular excessive menstrual bleeding and are found to have
infections, but is usually associated with other symptoms low haemoglobin levels should not donate blood and should be
BLOOD PRESSURE (BP) referred for medical assessment.
• A normal blood pressure (systolic 180 mmHg, diastolic 100 Contracting and relaxing the muscles in the legs, arms and abdomen
mmHg) is generally regarded as an indicator of good health. during donation may reduce the risk of vasovagal reactions,
particularly among female donors
The BTS should encourage donors to practise applied muscle
tension during blood donation

#MLSWATER2022 Page 13 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Accept female donors during menstruation, provided that they feel NOTE:
well and meet the minimum haemoglobin level for blood donation One unit of whole blood can be broken down into one unit of
Defer female donors during pregnancy and up to 6 months after packed red cells, one unit of platelets, and one unit of fresh
delivery or termination of pregnancy frozen plasma/cryoprecipitate.
Defer female donors during lactation

Reducing the risk of transfusion-associated acute lung injury (TRALI) Methods for collection of blood for preparation of blood components:
The gender of the donor may influence the type of blood Single whole blood donation
component prepared from the donation.
After collection of a unit of whole blood in the primary bag, blood
Plasma-rich blood components from multiparous women are
components can be separated from one another by differential
more likely to cause TRALI and related disorders than those from
centrifugation due to differences in their specific gravities.
males, because plasma from such women is likely to contain
After their separation, various components can be transferred from
alloimmune-reactive antibodies; these include antibodies to
one bag to another in a closed circuit thus avoiding exposure to the
human leucocyte antigens (HLA) or to human neutrophil antigens
external environment and maintaining the sterility.
(HNA), which are transferred passively during transfusion, to the
Blood should be processed for component separation within 6 hours of
possible detriment of a recipient who possesses the
collection
corresponding antigen.

Apheresis
Apheresis donation uses a specialized programmable machine that
Frequency of Donation processes over 3.5 L of the donors' blood and separates whole blood
The minimum interval between donations of whole blood should be into the blood component that is required (either up to 750 mL plasma
12 weeks for males and 16 weeks for females or 100–400 mL platelets), returning the remainder to the donor
The minimum interval between donations of platelets should be 4 (sometimes with saline fluid replacement).
weeks Depending on the component that is separated and removed, the
The minimum interval between donations of plasma should be 2 procedure is called plateletpheresis, leukapheresis, or plasmapheresis.
weeks Blood is withdrawn and returned via the 16-gauge apheresis needle
The minimum interval before an apheresis platelet or plasma (single arm/needle procedure).
donation should be 4 weeks following a whole blood donation, an Whole blood is mixed with anticoagulant and the required blood
apheresis red cell donation or a failed return of red cells during component (plasma or platelets) is collected into the attached plastic
apheresis blood bag.
In determining the frequency of donation and whether iron Red cell products can also be collected via apheresis, although this is
supplementation is given, the BTS should consider: less common than plasma and platelet apheresis.
• The need for longer donation intervals for young donors and Apheresis donation takes an average of 1 hour.
female donors of childbearing age Upon completion of the apheresis collection, the blood component
• Assessing the feasibility and affordability of providing iron requires no additional processing and has been depleted of the
supplementation to donors susceptible to donation-induced majority of white blood cells during the collection via filtration and/or
iron deficiency, especially women, adolescents, and repeat centrifugal conditions built in to the apheresis procedure.
and regular donors

#MLSWATER2022 Page 14 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Double red blood cell donation Directed or designated donation
A person donates twice as many red blood cells as with a single Family members or friends can donate blood specifically for one
donation of whole blood. another if the recipient's and donor's blood types and Rh factors are
This double donation is possible because the person gives only red compatible.
blood cells rather than whole blood. For some recipients, knowing who donated the blood is comforting,
Whole blood is drawn from the donor, and a machine that separates the although a donation from a family member or friend is not necessarily
blood into its components selectively removes the red blood cells and safer than one from an unrelated person.
returns the rest of the blood components (platelets and plasma) to the Blood from a family member is tested, as are all blood samples, and
donor. then treated with radiation to prevent graft-versus-host disease, which,
Some fluid is also given to the donor intravenously because otherwise, although rare, occurs more often when the recipient and donor are
the donor's blood pressure could become low enough to cause related.
symptoms, such as light-headedness or loss of consciousness.
After double red blood cell donation, people may be less able to Whole Blood
exercise vigorously for a few days. One unit of donor blood collected in a suitable anticoagulant-
Double red blood cell donation can be done as often as once every 112 preservative solution (citrate phosphate dextrose adenine or CPDA-1).
days (every 16 weeks). Whole blood donation involves collection of 450 mL (±10%) blood from
Some experts recommend that people take iron supplements after the antecubital vein via a 16- gauge needle into the attached specialized
double red cell donation so that their body can replace the donated red PVC plastic blood collection bag typically containing 63 mL
blood cells more rapidly. anticoagulant.
Whole blood donation usually takes 5–10 min.
Autologous transfusion The donated whole blood is then processed into components including
Donors are recipients of their own blood. red cell concentrate, platelet concentrate and plasma, via centrifugation
The person takes iron pills after donating the blood to help the body Processing of whole blood must commence within 24 hours of blood
replenish the lost blood cells before surgery. donation in order to maintain maximum quality of the components.
Also, during some types of surgery and in certain kinds of injuries, blood Whole blood is stored in an approved blood bank refrigerator at 1°-6°C.
that is lost can be collected, washed, and immediately given back to the If collected in ACD or CPD, shelf-life is 21 days and CPDA-1 is 35 days
person (intraoperative blood salvage). It does not contain functionally effective platelets and labile coagulation
An autologous transfusion eliminates the risks of incompatibility and factors (Factor V and Factor VIII).
blood-borne disease (unless the wrong blood is given by mistake). Transfusion of whole blood should commence within 30 minutes of
However, doctors do not use this technique as often as standard removal from the refrigerator and should be complete within 4 hours of
transfusion because the general blood supply is very safe due to starting.
rigorous donor screening and testing. Transfusion of one unit raises hemoglobin by 1 gm/dl or hematocrit by
In addition, older people may not tolerate donating blood befor surgery 3%.
because they are more likely to have side effects during donation such Indications
as low blood pressure and fainting. • Acute blood loss with hypovolemia
Older people are also more likely to have fewer blood cells than normal • Exchange transfusion in neonates
(a low blood count) to begin with. • Non-availability of red cell concentrate or suspension
Also, autologous transfusion is more expensive than standard
transfusion.

#MLSWATER2022 Page 15 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
APPENDICES Sample Problem # 2:
 Mathew, weighing 95 lbs, wants to donate blood for a friend. How
Sample Problem # 1: much must be removed from the blood bag?
 Given: 40kg individual that to donate a blood
a. Volume to Collect:  50 kg = 110 lbs
40 𝑘𝑔 a. Volume to Collect:
= 𝑥 450 𝑚𝐿
50 𝑘𝑔 95 𝑙𝑏𝑠
= 𝑥 450 𝑚𝐿
110 𝑙𝑏𝑠
 360 mL volume of blood to be collected
 388.64 mL volume of blood to be collected
b. Reduced Volume of Anticoagulant:
360 𝑚𝐿 d. Reduced Volume of Anticoagulant:
= 𝑥 63 𝑚𝐿
450 𝑘𝑔 388.64 𝑚𝐿
= 𝑥 63 𝑚𝐿
 50.4 mL 450 𝑚𝐿
 54.4 mL
c. Volume of Solution to be removed: e. Volume of Solution to be removed:
= 63 𝑚𝐿 − 𝑟𝑒𝑑𝑢𝑐𝑒𝑑 𝑎𝑚𝑜𝑢𝑛𝑡 𝑜𝑓 𝐴𝑛𝑡𝑖𝑐𝑜𝑎𝑔𝑢𝑙𝑎𝑛𝑡 = 63 𝑚𝐿 − 54.4 𝑚𝐿

 Answer: 12.6 mL  Answer: 8.6 mL

-END OF TRANSCRIPTION-

#MLSWATER2022 Page 16 of 16
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
MLS 18, April 13, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`
RATIO 8:1, 8 parts of the circulating blood & 1 part of the
Legend: anticoagulant
Transcription Bullet TRANSPORT 1°-10° C
Notes/Module Packet Bullet TEMPERATURE
 PPT SHELF-LIFE ACD, CPD, CPD2D: 21 DAYS,
CPD-A1: 35 DAYS,
MODULE OUTLINE CPDA-2/S: 42 days, (S indicated for additive sol’n.)
I. Organizations that Regulate or Accredit the Immunohematology Heparin 2 days/48 hours
Laboratory ADENINE- is related with the energy production, it
II. Blood Donor Selection increases the phosphorylation and ATP production in
III. Types of Donation order to provide the needed energy of our cells
IV. Blood Component
V. Safe Storage of Blood
Cellular Components
VI. Packing Blood Components for Transportation

LEARNING OUTCOMES Legend


At the end of this module, the student shall be able to:  Red cells
1. Identify the organizations that regulate or accredit the o Packed red cells
immunohematology laboratory. o Red cells in additive solution (Red cell suspension)
2. State the minimum acceptable levels for the following tests in o Leukocyte-poor red cells
allogenic & autologous donation o Washed red cells
3. Identify the medical history information that would cause o Frozen red cells
permanent deferral or temporary deferral and state the length of o Irradiated red cells
deferral period.  Platelets
4. Explain the reasons for permanent or temporary deferral given the o Platelet concentrate (Random donor platelets prepared
various medical conditions. from whole blood unit)
o Apheresis platelets (Single donor platelets) (SDP)
 Granulocytes
BLOOD COMPONENTS o granulocyte concentrate
Whole Blood
Cellular Components: Red Cells
INDICATION To increased volume and RBC mass, meaning that both PACKED RED CELLS
RBC mass/count and plasma volume is reduced or
decreased INDICATIONS:
STORAGE 1°-6° C is applicable to the whole blood and RBC • Anemia: Chronic severe anemia, severe anemia with
components congestive cardiac failure, anemia in elderly
#MLSWATER2022 Page 1 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
Do the heavy spinning at 5000 xg for 5 minutes in order to extract
INDICATION To increase RBC mass
platelet concentrate as well as the plasma
STORAGE 1°-6°C
TRANSPORT 1°-10°C LIGHT SPINNING
SHELF-LIFE Open system: 24 hours, without the use of centrifuge If you want a platelet concentrate and a packed RBC at the same
- Manually transfer the pocked RBC to the other time, then do the LIGHT SPINNING. 3200xg for 2-3 minutes and
side of the satellite bag thru the use of plasma the temperature for platelet is set at 20°-24° Celsius.
separator After light spin it will have a platelet rich plasma, from the
- Only viable for 24 hours platelet rich plasma, platelet concentrate can be separated from
Closed system: same as whole blood, commonly used the plasma
- Follows the shelf-life of the anticoagulant If you want to extract cryoprecipitate from the plasma, plasma
preservative that is used unit can be subjected to further centrifugation
DOSAGE Increase hemoglobin by 1g/dL
Increase Hematocrit by 3%
QC ≤ 80% of the Hct RED CELLS IN ADDITIVE SOLUTION (Red cell suspension)
• Acute blood loss (transfused along with a crystalloid or a INDICATIONS:
colloid solution) • Anemia: Chronic severe anemia, severe anemia with
congestive cardiac failure, anemia in elderly
Packed red cells are prepared by removing most of the plasma from • Acute blood loss (transfused along with a crystalloid or a
one unit of whole blood colloid solution)
Whole blood is either allowed to sediment overnight in a
refrigerator at 1-6°C or is spun in a refrigerated centrifuge. These are red cells with minimal residual plasma and an additive
Supernatant plasma is then separated from red cells in a closed solution (SAG-M which contains saline, adenine, glucose, and
system by transferring it to the attached empty satellite bag. mannitol).
Red cells and a small amount of plasma are left behind in the This increases shelf life from 35 days to 42 days.
primary blood bag. After collection of whole blood in the primary collection bag
Packed red cells have a high viscosity and therefore the rate of (containing CPDA-1), maximum amount of plasma is removed (after
infusion is slow. centrifugation) and transferred to one satellite bag.
Transfusion of one unit of red cells increases hemoglobin by 1 gm% The additive solution from the second satellite bag is transferred
(or increases hematocrit by 3%). into the primary collection bag (containing packed red cells) in a
closed system.

LEUKOCYTE-POOR RED CELLS


2 OPTIONS IN PREPARING PACKED RBCs
INDICATION To increased RBC mass as
HEAVY SPINNING
STORAGE 1°-6°C
If you want to prepare a packed RBC without platelet
TRANSPORT 1°-10°C
concentration, then do the HEAVY SPINNING with the whole
SHELF-LIFE Open system: 24 hours
blood sample.
Closed system: same as whole blood
Heavy spinning at 5000 xg for 5 minutes and set the temperature
DOSAGE Increase hemoglobin by 1g/dL
of the centrifuge at 4° Celsius.
#MLSWATER2022 Page 2 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
Increase Hematocrit by 3% Washed red cells are blood cells remaining after washing with saline
QC ≤ 85% RBC recovery, WBC should fall ≤5x10^6 solution to remove plasma
- Most commonly used blood component to o Open System
reduced HLA alloimmunization and CMV • needs to be transfused within 24hrs
transmission • can also be used w automated cell washers (Close
- the RBCs must be in patient suffering from System)
severe and recurrent febrile non-hemolytic
transfusion reaction. This is due to the FROZEN RED CELLS
presence of leukocyte antibody INDICATION Storage of rare blood and
autologous units
INDICATIONS: STORAGE -65 or -120°C
• Prevention of HLA immunization in patients who are likely to SHELF-LIFE 10 years
receive allogeneic bone marrow transplantation
• Prevention of febrile nonhemolytic transfusion reactions in If a cryoprotective agent such as glycerol is added, red cells can be
persons receiving multiple transfusions stored frozen for up to 10 years.
• Prevention of transmission of cytomegalovirus. This method can be used for storage for donor red cells with rare
blood groups, for future autologous transfusion, and for individuals
Leukocyte-poor red cells contain < 5 × 106 white cells per bag. who have repeated febrile nonhemolytic transfusion reactions.
Methods for leukocyte depletion are: used in the long-term storage of rare blood units for
• Leukocyte-reduction filters autologous units and units for special purpose in cases of the
• Removal of buffy coat intrauterine transfusion
In Frozen RBC, one cannot directly transfuse frozen RBC in the
WASHED RED CELLS patient.
INDICATION To increased RBC mass in patient Cryoprotective agent classifications
with allergic, anaphylactic, febrile o Penetrating Agent
and urticarial reactions  small molecules
STORAGE 1-6°C  can enter the cell and prevent cell dehydration as the ice
SHELF-LIFE Open system: 24 hours forms; ex. Glycerol & DMSO
DOSAGE Increase Hb by 1g/ dL o Non-penetrating Agent
Increase Hct by 3%  includes Hydroxyethyl starch, glucose, polyvinyl-povidone
QC ≤ 70% Hct  large molecules; does not enter the cell
 forms shell around the cell thereby preventing the loss of
Red cells can be washed with normal saline to remove plasma
water
proteins, white cells, and platelets.
Prepared by washing packed RBC by multiple batch processing THREE METHODS IN FREEZING
through the process of centrifugation and decanting 1. High Glycerol Method
supernatant saline solution uses 40% of weight per volume of the final concentration
Such red cells are used for IgA-deficient individuals who have To slow uncontrolled freezing and most common method used
developed anti-IgA antibodies, as exposure will lead to anaphylaxis. RBC units can be stored at -80C using mechanical reader
#MLSWATER2022 Page 3 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
2. Low Glycerol Method Whole blood can be pulled into batches of 6-8 units prior to
uses 20% of weight per volume transfusion
more rapid and controlled compared to High Glycerol
PRP is then transferred to the attached satellite bag and spun (high
freezing temp is much lower at -120C and uses liquid nitrogen
spin) to get platelets at the bottom and supernatant plasma.
freezer Most of the supernatant is returned back to the primary collection
3. Agglomeration bag or to another satellite bag, leaving behind 50-60 ml of plasma
employs deglycerolization technique in which the presence of with the platelets.
low ionic strength solution, the cells will agglomerate, forming Platelets are stored at 20°-24°C with continuous agitation (in a
large clumps that will sink at the bottom of the bag storage device called platelet agitator).
o the supernatant can be removed and wash procedure can 22-24 deg C; Units = 5.5x1010 platelets, must contain
be reinstituted sufficient plasma to maintain pH =  6.2

Maximum period of storage is 5 days.


IRRADIATED RED CELLS Transfusion of one unit will raise the platelet count in the recipient
INDICATION Prevent TA-GVHD by about 5000/μl.
RADIATION SOURCE Cesium (137 Cs), Cobalt (60 Co) 1 RBC unit can increase blood platelet count by 5,000 to 10,000
SHELF-LIFE 28 days from irradiation or per uL in a typical 70g human
original expiration date
The usual adult dose is 4-6 units of platelet concentrate (or 1
unit/10 kg of body weight).
Gamma-irradiation of red cells inactivates lymphocytes and These units (which are from different donors) are pooled into one
prevents graft vs. host disease. bag before transfusion.
Irradiated red cells are indicated for intrauterine or premature This dose will raise the platelet count by 20,000 to 40,000/μl.
neonate transfusions, and in individuals with immunodeficiency,
and in those receiving blood from first-degree relative donors.
APHERESIS PLATELETS (Single donor platelets)
In platelet pheresis, a donor is connected to a blood cell separator
Cellular Components: Platelets machine in which whole blood is collected in an anticoagulant
INDICATIONS: solution, platelets are separated and retained, and remaining
• Bleeding due to decreased platelet production components are returned back to the donor.
• Bleeding in hereditary disorders of platelet function With this method, a large number of platelets can be obtained from
• Massive blood transfusion a single donor (equivalent to 6 units of platelet concentrate).
PLATELET CONCENTRATE (Random donor platelets prepared from This method is especially suitable if HLA-matched platelets are
whole blood unit) required (i.e. if patient has developed refractoriness to platelet
Platelet concentrates can be obtained from single donor units or by transfusion due to the formation of alloantibodies against HLA
plateletpheresis. antigens).
One unit of whole blood is centrifuged (light spin) to obtain platelet-
rich plasma (PRP). Through the use of apheresis
Whole blood must be processed within 6-8hrs after collection 22-24 °C; Shelf life = 5 days

#MLSWATER2022 Page 4 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
11
Units = 3.0 x 10 platelets per unit; equivalent to 6-10 units of FFP- Plasma units that were prepared within 6hrs after
random donor platelet collection with ACD as anticoagulant, within 8hrs if CPD or
Used to increase the platelet count from 30,000 to 60,000 uL CPDA1 was used
Why do we need constant Agitation? Plasma is separated from whole blood by centrifugation, expressed
 That is to maintain maximum biological function. into the attached satellite bag, and rapidly frozen at –20°C or at
 To prevent clotting. lower temperature.
 To prevent clumping of platelets. FFP contains all the coagulation factors.
FFP can be stored for 1 year if temperature is maintained below –
 For the oxygen to be sufficient in the storage.
25°C.
 To facilitate oxygen transfer into the platelet bag and oxygen
Stored at -80°C for 1 year
consumption.
If wanted to prolong shelf life, it can be stored in a much
lower temperature
o Usually: -65 °C
Cellular Components: Granulocytes
Granulocyte Concentrate When required for transfusion, FFP is thawed between 30- 37°C
and then stored in the refrigerator at 2-6°C.
Plasma Components must be placed in a plastic bag to prevent any
Legend contamination
 Fresh frozen plasma (FFP) Since labile coagulation factors rapidly deteriorate, FFP should be
 Cryoprecipitate transfused within 2 hours of thawing.

Fresh frozen plasma (FFP) Plasma Frozen 24


INDICATIONS: Plasma Frozen within 24 hours or PF24
• Multiple coagulation factor deficiencies: liver disease,
warfarin overdose, massive blood transfusion Cryoprecipitate
• Disseminated intravascular coagulation
• Inherited deficiency of a coagulation factor for which no INDICATION Fibrinogen deficiency, hemophilia
specific replacement therapy is available a, vWF disease, Factor XIII
• Thrombotic thrombocytopenic purpura deficiency
STORAGE ≤ -18
Plasma collected from platelet concentrates or from the platelet CONTENT Fibrinogen: 150 mg
rich plasma preparation can either be prepared as: AHF: 80 IU
1. FFP or Fresh Frozen Plasma vWF, FXII
2. Plasma Frozen 24 SHELF-LIFE 1 year
NOTES White mass of precipitate in 15
FFP or Fresh Frozen Plasma mL plasma
FFP is prepared from whole blood within 6 hours of collection
because after this time labile coagulation factors are lost. INDICATIONS:
• F VIII deficiency (if F VIII concentrate is not available)
#MLSWATER2022 Page 5 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
• Von Willebrand disease Thawed units cannot be refrozen
• Deficiency of fibrinogen.

Cryoprecipitate is prepared from plasma that has been freshly Plasma Derivatives
separated (within 6 hours of collection) by rapidly freezing it at - Legend
20°C or lower and thawing it slowly at 4-6°C.  Human albumin solutions
A white flocculent precipitate and plasma are obtained.  F VIII concentrate
The mixture is centrifuged and supernatant plasma is removed  F IX concentrate
leaving behind sediment of cryoprecipitate suspended in 10-20 ml  Prothrombin complex concentrate (PCC)
of plasma.  Immunoglobulins
The unit is then refrozen (-20°C or colder) and can be stored at this o Non-specific immunoglobulins
temperature for 1 year. o Specific immunoglobulins
When needed, cryoprecipitate is thawed at 30-37°C, required Plasma derivatives are manufactured by fractionation of large
donations are pooled and transfused to the patient. volumes of pooled human plasma.
Cryoprecipitate contains
• F VIII
• von Willebrand factor Human albumin solutions
• fibrinogen Albumin is prepared by cold ethanol fractionation of pooled
• F XIII plasma and is sterilized during manufacture to destroy viruses and
• fibronectin. bacteria.
Albumin is used as a replacement fluid in therapeutic plasma
Prepared from FFP, allow FFP to thaw then do thawing in a slower exchange, and for treatment of diuretic-resistant edema of
method hypoproteinemia.
Place FFP in a refrigerator at 1-6 °C in order to allow the ice formed
crystal in FFP to cool down Both the normal serum albumin and plasma protein fractions are
o Usually takes hours 14-16 hours in slow thawing used in treatment of hypo bulimic and hypo proteinimic cases and
In using cryoprecipitate thaw bath set at 4 °C it will only take 4 patients who are in shock or have severe burns
hours to cool down Have higher amount of Albumin than TPF, 96% albumin and 4%
o Endpoint of thawing is when presence of slushy precipitate globulin.
is present in plasma. Stored at 2-10 °C for 5 years
o Centrifuge using hard spin to separate the supernatant Granulocyte products used to treat severe neutropenia and
plasma from the precipitate you leave approximately 10- granulocyte dysfunction. Stored 20-24 °C for 24 hours
15ml of the plasma on the precipitate is now the
Cryoprecipitate
Freeze within 1 hour from the time the plasma reaches the slushy F VIII concentrate
state.
Supernatant fluid can be refrozen and label as cryo-poor plasma. INDICATION Prevents or control bleeding in
Thawed unit must be kept at room temperature until transfusion. If Hemophilia A
several bags of precipitate are pulled blood must be infused or STORAGE 1-6°C
transfused within 4 hours
#MLSWATER2022 Page 6 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
Non-specific immunoglobulins:
Freeze-dried F VIII concentrate is prepared by fractionation from INDICATIONS:
large pools of fresh frozen plasma. • passive prophylaxis of viral infections like hepatitis, rubella,
To reduce the risk of transmission of viral infections, it is treated and measles
with heat or chemicals during manufacturing process. • treatment of hypogammaglobulinaemia,
F VIII concentrate is the treatment of choice for treatment of • autoimmune thrombocytopaenic purpura to induce a rise
hemophilia A and severe von Willebrand disease. in platelet count
• neonatal sepsis
Specific single factor concentrates were also used in treatment of
diseases These are derived from the pooled plasma of non-selected
factor VIII used in treatment for hemophilia A and factor IX for donors.
hemophilia
Usual factor concentrates were VIII, IX, XIII are actually lyophilize
Specific immunoglobulins:
products prepared from pulled plasma using con ethanol method.
They are obtained from donors who have selected high titer IgG
o Lyophilization must be in order to prevent transmission of
antibodies.
viruses
Anti-RhD immunoglobulin is prepared from plasma of Rh-
Pasteurization method, solvent and detergent method, monoclonal
negative donors who have produced anti-D following
purification method, and ultraviolet irradiation method
immunization; it is used for prevention of sensitization to RhD
antigen in Rhnegative women giving birth to a Rh-positive baby.
F IX concentrate Other specific immunoglobulins include hepatitis B immune
globulin, varicella-zoster immune globulin, and tetanus immune
Prothrombin complex concentrate (PCC) globulin that are used for passive prophylaxis of infections.
MAIN USES/ INDICATIONS:
• Deficiency of F IX
• Deficiency of F VIII with development of inhibitors against F
VIII SAFE STORAGE OF BLOO D
• Inherited deficiency of factors II, VII, and X. Whole blood
Whole blood and red cells must always be stored at a temperature
PCC contains factors II, VII, IX, and X, and also protein C and S. between +1°C and +6 °C.
A serious risk of PCC is thrombotic complications due to the The main reasons for giving a blood transfusion are to restore or
presence of small amounts of activated coagulation factors. help to maintain the body’s oxygen-carrying capacity and the
volume of blood circulating around the body.
Immunoglobulins • If blood is not stored at between +1 °C and +6 °C, its oxygen-
carrying ability is greatly reduced.
Immunoglobulins are obtained by cold ethanol fractionation of large
• The anticoagulant/preservative solution in the blood bag
pools of human plasma.
contains nutrients for the blood during storage and stops the
They are of two types: specific and nonspecific.
blood from clotting.

#MLSWATER2022 Page 7 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
• The red cells can only carry and deliver oxygen if they remain Cryoprecipitate
viable: that is, if they retain the same properties as they have Cryoprecipitate is the cold insoluble portion of plasma remaining
during their normal circulation in the body. after FFP has been thawed between +1 °C and +6 °C and is useful for
Another important reason for storing blood between +1 °C and correcting certain coagulation defects.
+6 °C is to keep the growth of any bacterial contamination in the It contains approximately 50% of Factor VIII and von Willebrand
unit of blood to a minimum. Factor, 20–40% of fibrinogen and some of the Factor XIII originally
• If blood is stored above +6 °C, bacteria that may have present in the fresh plasma.
inadvertently entered the unit during collection may grow to Plasma is separated from red cells within 6 to 8 hours of collecting
such an extent that transfusion of the contaminated blood blood.
could be fatal. The plasma is frozen solid rapidly, certainly within 30 minutes of
The lower limit of +1 °C is also very important. separation from the cells.
• This is because red cells are very sensitive to freezing. The plasma is then thawed slowly at below +4 °C.
• If they are allowed to freeze, the red cell membranes rupture In order to get the maximum yield of Factor VIII in the
and the haemoglobin is released; that is, the cells are cryoprecipitate from a blood unit it is important to adhere strictly to
haemolysed. the standard procedures for the collection, storage and processing
• The transfusion of haemolysed blood can also be fatal. of the component.
The stability on storage is dependent on the storage temperature
Fresh frozen plasma available.
Fresh frozen plasma (FFP) is plasma that has been separated from a • The optimal storage temperature is below –30 °C.
unit of whole blood within 6 to 8 hours of collection, and has been
rapidly frozen and maintained at all times at a temperature of – Platelet concentrates
20 °C or lower. Platelet transfusions are used to prevent spontaneous bleeding or
There is no lower temperature limit for the storage of FFP, to stop bleeding in patients with established thrombocytopenia or
although the optimal temperature is –30 °C or lower platelet dysfunction
Plasma contains water, electrolytes, clotting factors and other • Ex., hypoplastic anemia or bone marrow failure – due to
proteins (mostly albumin), most of which are stable at refrigerator replacement with malignant cells or to the effects of
temperature, i.e. +1 °C to +6°C. chemotherapy.
Factor V and Factor VIII, however, which are essential in the clotting Both manual and automated methods can be used in the
mechanism, will deteriorate and diminish in quantity if they are not preparation of platelet concentrates.
stored at –20 °C or lower and greatly reduce the clotting activity of Lower temperatures adversely affect platelet function and viability.
the plasma. • For this reason, whole blood should be kept at between
FFP may be given to a patient to restore or help to maintain +20 °C and +24 °C until it is processed into platelet
coagulation factors such as Factor V or Factor VIII. concentrates and other blood components.
Plasma should not be used as a volume expander unless crystalloids Platelet-rich plasma must be separated from whole blood by
and colloids are unavailable centrifugation within 8 hours of phlebotomy.
Additional centrifugation and removal of most of the supernatant
plasma may then concentrate the platelets.
Platelet concentrates should be stored at a temperature of between
+20 °C and +24 °C with continuous agitation.

#MLSWATER2022 Page 8 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
• This is essential to prevent platelet aggregation which results Plasma derivatives
in loss of viability. Unlike blood components, plasma derivatives such as albumin or
The shelf life and transport conditions differ according to the type of immunoglobulin are concentrated, sterile specific proteins,
plastic bag used to store the component. obtained from large pools of donor plasma through a complex
Platelet concentrates stored at between +20 °C and +24 °C maintain pharmaceutical process called plasma fractionation.
their function and viability better than refrigerated platelet They are used to treat patients with specific protein deficiencies or
concentrates. requirements for passive immunity.
Current plasticizers used in the manufacture of plastic bags allow for
storage of up to five days, because gaseous exchange takes place PACKING BLOOD COMPONENTS FOR TRANSPORTAT ION
between the container and the environment and this results in the The following general observations must be kept in mind:
maintenance of pH in the component, which is critical for platelet
storage. Label the container THIS WAY UP with an arrow.
Ice should be placed above the blood because cool air moves
Note:
downwards.
If no platelet agitator or rotator is available, it is not possible to store
o Cubed wet ice may be better than chipped or broken ice for
platelets.
long distance shipments of blood because it melts more
Once prepared, they must be transfused immediately unless the
slowly.
blood bank is equipped with:
o Ice packs can be used at –5 °C or below.
an air-conditioned facility with a temperature monitoring system
The recommended storage conditions must be maintained when
that will maintain an ambient temperature of between +20 °C
blood is moved from one location to another, including:
and +24 °C or
o from a mobile or satellite collection site to the laboratory
- a platelet incubator that will keep the platelet concentrates at a
o from the blood bank to a different facility (to a hospital or
temperature of between +20°C and +24 °C.
clinic or another blood bank)
o from the blood bank to hospital wards or operating rooms
Since platelet concentrates are stored at room temperature, they
pose a greater risk for bacterial proliferation.
SOPs on the cleaning of the venipuncture site prior to donation Red cell components:
must be strictly followed, and the disinfectant in use must undergo at no point should ice be allowed to come into direct contact with
regular quality control checks. the blood as the red cells nearest to the ice may freeze and
hemolyze.
Storage conditions and expiry dates should also be strictly adhered o In boxes shipped long distances or at high environmental
to in order to prevent septic shock for the recipient. temperatures, the volume of ice should at least equal that
After the hermetic seal is broken, platelet concentrates should be of the blood.
transfused as soon as possible, but definitely within a maximum of 4 o In an insulated container, the temperature can be
hours of storage at between +20 °C and +24 °C. considered to be in the +2 °C to +10 °C range as long as
unmelted ice is still present on arrival at destination.

Plasma
There should be at least as much wet ice in the cold box as there is
plasma.

#MLSWATER2022 Page 9 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 2)
o It is important to protect the frozen plasma units during
transportation.
o If possible, they should have been placed in cardboard
boxes before freezing to protect the bags from developing
small cracks.
o A simple method to determine if plasma units have thawed
and refrozen is to place a rubber band around the unit at
the time of preparation.
o Once the unit freezes it leaves an indentation at the sides.
o If the unit has thawed, or thawed and refrozen, the
indentation will not be there.

Platelets
Containers for transporting platelets should be equilibrated at a
temperature of +20 °C to +24 °C before use.
o If outdoor temperatures are extremely high, special
chemical, coolant pouches are available that may be
shipped with platelets and will maintain temperatures of
approximately +20 °C to +24 °C for up to 12 hours.
o Also available are containers with a power source that
maintains temperatures between +20 °C and +24 °C.
o Platelets should reach their destination within 24 hours,
which is the maximum time allowed without agitation.

-END OF TRANSCRIPTION-

#MLSWATER2022 Page 10 of 10
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 3)
MLS 18, April 6, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`

Legend:
Transcription Bullet 14. State the expected incremental increase of a
Notes/Module Packet Bullet a) patient’s hematocrit level following transfusion of each unit of RBCs
 PPT and
MODULE OUTLINE b) platelet count following transfusion of each unit of platelet.
15. Identify the group of recipients who are at high risk of infection from
I. Test required for donations
transfusion of cytomegalovirus-positive RBcs or platelets.
II. Therapeutic Apheresis
16. Explain the role of irradiation in the prevention of tansfusion-
III. Transfusion Therapy
associated graft- versus-host disease
17. State the respective blood products of choice for treating von
MODULE OUTLINE Willebrand’s disease, hemophilia A, and hemophilia B
At the end of this modules, the student shall be able to: 18. List the main advantages and disadvantages of autologous
1. Differentiate among the four different methods of autologous blood transfusion.
donation. 19. Identify the most important factors to consider when emergency
2. Describe the procedure for a whole blood donation and post transfusion is indicated.
phlebotomy care instructions for the donor. 20. Define massive transfusion.
3. Differentiate mild, moderate, and severe donor reactions and list
recommended treatments for each.
4. List the tests required for allogenic, autologous, and apheresis
TESTS REQUIRED FOR DONATIONS
donortions.
5. State the acceptable interval of donation for allogenic and apheresis ABO/Rh
donors. Testing for the donor’s ABO group must include both forward and
6. List the labeling criteria for a unit allogenic and autologous blood. reverse grouping.
7. Define apheresis; leukapheresis, platelepheresis, plamapheresis, & The ABO group must be determined by testing the donor RBCs with
erythcypheresis. anti-A and anti-B reagents and by testing the donor serum or
8. Compare and contrast the procedures of continuous flow plasma with reagent A1 cells and B cells
centrifugation and intermittent flow centrifugation. The donor’s Rh type should be determined by testing with anti-D
9. List the components that can be collected using apheresis technology. reagent at the immediate spin phase.
10. Explain the rationale for the basic types of therapeutic apharesis.
11. Explain the adverse effects of apharesis. Antibody Screen
12. Describe the blood products that are currently available for Donors with a history of pregnancy or transfusion must be tested
therapeutic us - the composition and approximate volume of each. for unexpected antibodies to RBC antigens
13. Select the appropriate blood product for patient with specific Uses a pooled cell reagent:
disorders. o This reagent contains two or three individual cells pooled
into a single reagent.
#MLSWATER2022 Page 1 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
o The pooled screening cell reagent contains all of the Deferral
required antigens and is capable of detecting the presence  A positive result for anti-HBc along with a positive HBsAg would place
of the clinically significant alloantibodies. the donor in a permanently deferred category.
The antibody screen performed on a recipient prior to transfusion  A positive test for anti-HBc without an accompanying positive test for
uses two or three individual reagent cells HBsAg or HBC is not cause for donor deferral unless it occurs on more
than one occasion or on two consecutive tests.
HBsAg o The products will not be used for transfusion even if the donor is not
Methods currently available include: deferred for a positive result
o Enzyme immunoassay (EIA)
o Chemiluminescence (ChLIA)
o Nucleic acid amplification (NAT) Anti-HCV
Is able to detect small amounts of viral nucleic acid in blood before
Deferral antibodies or viral proteins such as HCV core antigen are detectable
 If the EIA or ChLIA tests are initially positive, they must be repeated by current methods.
in duplicate and a confirmatory (neutralization) test must be Screening tests for anti-HCV involve:
performed. o EIA
o If the confirmatory test is positive, the donor is considered to be o ChLIA
infected (acute or chronic) with the hepatitis B virus (HBV) and must o NAT
be permanently deferred Confirmatory methods include:
 NAT is not mandated for donor screening o RIBA (recombinant immunoblot assay)
o If the initial HBsAg testing is reactive but the confirmatory testing is o HCV RNA
not (unconfirmed positive), all the current products are discarded
but the donor need not be permanently deferred, provided the Deferral
antibody to hepatitis B core (anti-HBc) testing is also nonreactive.  An individual who is positive by RIBA is considered to have the
o The donor is deferred for 8 weeks and may be reinstated if the next HCV antibody and would be indefinitely deferred as a blood
HBsAg testing is nonreactive. donor
 Donors who test repeat reactive for HCV screening tests must be
deferred, and the components or products prepared are
Anti-HBc discarded.
The methods employed are similar to those for HBsAg.  If the confirmatory RIBA or NAT testing are nonreactive, the
Presence of HBc antibody in the donor serum suggests the donor may be considered for reentry.
possibility of HBV infection, either acute or chronic.

Anti-HIV-1/2
All donor units must be screened for the presence of the human
immunodeficiency virus (HIV-1/2) antibody using an FDA-approved
method.

#MLSWATER2022 Page 2 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
o If the initial screening test is negative, the unit is suitable for Any individual units that test reactive should be discarded, and the
transfusion; donor should be deferred from further donations for 120 days
o If it is positive, the test must be repeated in duplicate.
Screening tests include: Syphilis
o EIA A serologic test for syphilis is done because the disease is
o ChLIA characterized as being sexually transmitted and places the donor at
o NAT (run using 16 to 24 donation samples per pool) higher risk for possible exposure to hepatitis and HIV
Confirmation tests for HIV include: Screening tests include:
o Western blot (Wb) o Rapid plasma regain (RPR)
o Immunofluorescence assay (IFA) o Venereal Disease Research Laboratory (VDRL).
o Results are expressed as positive, negative, or indeterminate o Both tests are based on Reagin, or antibody directed toward
cardiolipin particles.
Anti-HTLV-I/II o Cardiolipin-like antibodies have been documented in
HTLV-I virus or human T-cell lymphotrophic virus type I is the persons with untreated syphilis infections.
causative agent of adult T-cell leukemia o Antibody will agglutinate cardiolipin carbon particles in the
Associated with a neurological disorder called HTLV-associated form of visible flocculation.
myelopathy. The confirmatory test for syphilis is:
HTLV-II has been shown to have about 60% homology with type I o FTA-ABS or fluorescent treponemal antibody absorption
and is prevalent among intravenous drug users in the United States. test
Persons can contract both viruses from transfusion via infected o Indirect immunofluorescence is used to detect antibodies to
lymphocytes the spirochete T. pallidum, the agent that causes syphilis
Screening test methodologies include: o If the STS or VDRL screening test is reactive or
o EIA indeterminate, the components from that donation must be
o ChLIA discarded and the donor deferred unless a confirmatory
Confirmatory tests include: FTA-ABS test is nonreactive.
o Western blot o If the FTA-ABS test is nonreactive, the donor may be
o RIPA reentered and the components labeled as reactive with the
o NAT testing screening test.
o The donor of a confirmed reactive test may be reentered
WNV RNA into the donor pool after 12 months and documentation of
Test include: completion of treatment
Mini-pool (MP-NAT)
o Uses pools of 6 to 16 donor samples T. cruzi (Chaga’s Disease)
o Is routinely used when risk of WNV infection in the Chagas’ disease is a parasitic infection that is endemic to Mexico
geographical area is low and Central and South America
The infection is generally mild but persists, without symptoms,
Individual donor (ID-NAT) method throughout the infected individual’s life and can be transmitted
o Recommended when the risk is high through blood transfusions
All donors (allogeneic and autologous) are to be tested once.

#MLSWATER2022 Page 3 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
o If the results are nonreactive, the donor need not be to remove a particular blood component for therapeutic purposes
retested with each donation. (therapeutic apheresis).
o This is because most donors who are infected in the United The process of removing plasma is termed plasmapheresis. In a
States have a chronic infection that was acquired when similar manner, platelets (plateletpheresis), red blood cells
residing in a country where the disease is endemic. (erythrocytapheresis), or leukocytes (leukapheresis) can be
o Repeat reactive donors must be deferred indefinitely and removed (or collected) using apheresis technology.
the products quarantined and destroyed Citrate is used as the primary anticoagulant in apheresis procedures.
o The binding of calcium ions by citrate inhibits the calcium-
Platelet Bacterial Detection dependent coagulation cascade
Platelet components are particularly at risk because they are stored o Citrate is mixed with the blood immediately as it is removed
at room temperature from the donor’s (or patient’s) vein, and effectively
For apheresis-derived platelets, it is recommended that a culture anticoagulates the blood before it enters the apheresis
method be used that would usually be performed by the collecting machine
facility. The variables that are considered during an apheresis procedure
For whole blood–derived platelets, there are two choices. include:
For platelets that are pooled within 4 hours of transfusion, a new o Centrifuge speed and diameter
PGD test (Verax) can be used. o Duration of dwell time of the blood in the centrifuge
o It is a relatively easy test to perform, requires about 45 o Type of solutions added, such as anticoagulants or
minutes, and can be done by a transfusion service at the sedimenting agents
time of issue. o Cellular content or plasma volume of the patient or donor
For platelets that are pooled and stored as a pre-pooled product, a
culture-based test is recommended. Methods of Centrifugation
o Typically performed by the collection facility
INTERMITTENT FLOW CENTRIFUGATION (IFC)
Blood is processed in batches or cycles
PROCEDURES USED TO SEPARATE BLOOD COMPONENTS
Whole blood is drawn from an individual with the assistance of a
Apheresis pump
A procedure in which whole blood is removed from the body and To keep the blood from clotting, an anticoagulant is mixed with the
passed through an apparatus that separates out one (or more) blood as it is pumped into a centrifuge bowl through the inlet port
particular blood constituent. Can be performed as a single-needle procedure with only one
Returns the remainder of the constituents to the individual’s venipuncture
circulation. This is advantageous when collecting apheresis products from blood
It allows a larger volume of specific components to be collected donors.
It permits the removal of disease-causing or unwanted cellular or If both arms are used (double-needle procedure: one for
plasma constituents from a patient. phlebotomy and one for reinfusion), the amount of time for the
It is used to harvest stem cells from the peripheral blood of donors apheresis can be reduced
and patients, avoiding the need for extraction from the bone Procedure involved complete a cycle before beginning the next one
marrow Usually smaller and more mobile
Apheresis can be performed on a donor to collect a specific blood A single venipuncture may be used
component (donor apheresis), or it can be performed on a patient

#MLSWATER2022 Page 4 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
CONTINUOUS FLOW CENTRIFUGATION (CFC) RED BLOOD CELLS
The processes of blood withdrawal, processing, and reinfusion are RBCs collected by apheresis are typically collected as a double unit
performed simultaneously in an ongoing manner. (termed a 2RBC or double RBC procedure)
Because blood is drawn and returned continuously during a A clinical advantage to the collection of apheresis RBCs is reduced
procedure, two venipuncture sites are necessary donor exposure for the recipient since the patient can potentially
Separation of the components is performed by centrifugation, and receive two units from the same individual
the specific component is diverted and retained in a collection bag. Since the volume of RBCs being collected during a 2RBC procedure
The remainder of the blood is reinfused to the individual via the is greater than it would be for a whole blood donation, the
second venipuncture site. requirements for donor hematocrit are more stringent.
Two venipunctures are usually required o The hematocrit must be at least 40% regardless of gender,
and the level (hemoglobin or hematocrit) must be
Membrane filtration technology can also be used to separate determined by a quantitative method;
blood components o The use of copper sulfate is not acceptable
Membrane separators are typically composed of bundles of hollow If two units of RBCs are collected by apheresis, the donor must wait
fibers or flat plate membranes with specific pore sizes 16 weeks before providing another donation that includes RBCs.
This technology lends itself well to the collection of plasma, since If one RBC and one plasma and/or platelet unit are collected, the
pores can be sized to prevent the passage of even small cellular donor must wait 56 days before donating another red cell product
elements.
Filtration has several advantages: PLASMA
o The collection of a cell-free product In a plasmapheresis procedure, whole blood from the donor is
o The ability to selectively remove specific plasma proteins centrifuged, the plasma is diverted into a collection bag, and the
by varying the pore size cellular components (RBCs, platelets, WBCs) are returned to the
donor.
COMPONENT COLLECTION o This allows a larger volume of plasma to be collected from a
A qualified, licensed physician must be responsible for all aspects of donor, such that each apheresis unit (“jumbo” plasma) is
the apheresis program the volume equivalent of at least two whole-blood-derived
Operators of apheresis instruments may include: plasma units.
o Medical technologists (clinical laboratory scientists) Purposes:
o Nurses o It can be used to augment the inventory of fresh frozen
o Technicians trained on the job plasma (FFP) of a particular ABO group, especially group AB
Written, informed consent must be obtained from the donor o To prepare immune globulin to provide prophylaxis against
The apheresis procedure is more comfortable for the donor: infectious organisms in exposed individuals
o Since the needle used for venous access is typically of a o Is used commercially to collect plasma for further
larger gauge (i.e., smaller size) than for whole blood manufacturing into such products as intravenous immune
collection globulin (IVIG), hepatitis immune globulin, and Rh-immune
o The infusion of saline during the procedure helps reduce globulin.
donor reactions due to hypovolemia With infrequent plasmapheresis, donation occurs no more than
once every 4 weeks, and the donor requirements are the same as
for whole blood

#MLSWATER2022 Page 5 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
With frequent, or serial, plasmapheresis, donation occurs more o To protect the donor, if a double or triple apheresis platelet
frequently than once every 4 weeks is collected, 7 (rather than 2) days must elapse before the
o There must be at least 2 days between procedures and no donor is again eligible to provide apheresis platelets
more than two procedures in a 7-day period o A donor may undergo no more than 24 plateletpheresis
procedures in a rolling 12-month period
PLATELETS The total volume of plasma collected during any one procedure
Platelets obtained by an apheresis procedure provide the equivalent cannot exceed 500 mL (or 600 mL if the donor weighs 175 lb or
of six to eight whole-blood-derived platelets (random-donor more) or the volume of plasma cleared by the FDA for the
platelets) instrument
o This significantly decreases the donor exposure for a patient
In a plateletpheresis procedure, platelets along with a portion of GRANULOCYTES
plasma are removed, and the remaining RBCs, WBCs, and majority Granulocyte transfusions have been shown to be beneficial in some
of the plasma are returned to the donor severely neutropenic patients (neutrophil count less than 500/µL)
o The platelets are suspended in donor plasma in a collection who meet the following criteria:
bag specifically designed for platelet storage o Documented (clinically or by culture) infection for 24 to 48
A routine plateletpheresis procedure typically takes 45 to 90 hours that is unresponsive to standard antibiotic or
minutes antifungal therapy
Donor selection criteria for the plateletpheresis donor are the same o Bone marrow demonstrates myeloid hypoplasia
as for whole blood donation, with two additional requirements o There is a reasonable chance of bone marrow recovery (i.e.,
o Prior to each plateletpheresis procedure, a sample must be the neutropenia is reversible)
collected to determine the donor’s platelet count. Granulocyte transfusions have also shown favorable results in the
o The platelet count must be at least 150,000/µL in order to treatment of neutropenic neonates with sepsis
provide an adequate platelet collection and for the donor to Collection of granulocytes by apheresis provides a higher yield
safely undergo the collection procedure. product
o If the donor’s platelet count is less than 150,000/µL, he or Apheresis collection requires close communication between the
she is deferred from platelet donation until a subsequent blood center or apheresis center, the blood bank physician, and the
count is at least 150,000/µL patient’s physician
Plateletpheresis collections should not be performed on potential A minimum therapeutic dose is 1 × 10¹⁰ granulocytes per day
donors taking medications that interfere with platelet function, as The granulocyte yield is influenced by the donor’s neutrophil count
this would result in production of a suboptimal and therapeutically and the collection process
ineffective patient product During centrifugation of whole blood, granulocytes are found in the
Antiplatelet medications have differing deferral time periods: buffy coat between the RBC and plasma layers (see Fig. 14–2).
o 48 hours for aspirin, aspirin-containing medications, and the Adding the red cell sedimenting agent, hydroxyethyl starch (HES),
anti-inflammatory drug Feldene allows better separation of layers, resulting in an improved yield
o 14 days for clopidogrel (Plavix), ticlopidine (Ticlid), ticagrelor with reduced RBC contamination.31,32 Since HES is added directly
(Brilinta), and prasugrel to the apheresis circuit, some amount will enter the donor’s
The interval between plateletpheresis procedures must be at least 2 circulation during the collection procedure and ultimately be
days with no more than two procedures in a 7-day period removed by the reticuloendothelial system. Immediate side effects

#MLSWATER2022 Page 6 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
of HES are due to its colloid properties, including circulatory volume
expansion, with headaches and peripheral edema Adequate vascular access is mandatory during TA, as larger volumes
of blood are processed and the duration of the procedure is longer
THERAPEUTIC APHERESIS (TA) than for donor apheresis
Like donor apheresis, this involves the removal of a specific blood
component, with return of the remaining blood constituents to the Vascular access can be obtained via:
patient  Peripheral veins
o However, with TA, since the component being removed is o If only one to two TA procedures are to be
considered pathological (or contributing to the patient’s performed, peripheral access can be used
underlying disease state), significantly larger volumes of o In such instances, two venous sites are necessary—
blood must be processed in order to remove as much of the one for removal and one for return
offending agent as possible  Central veins
The TA procedure is classified according to the blood component  A combination of both
removed:
o A cytapheresis procedure may be used to selectively Therapeutic plasma exchange (TPE) is the removal and retention of
remove RBCs, WBCs, or platelets the plasma, with return of all cellular components to the patient
o A plasmapheresis procedure is used to remove plasma o This is the most common TA procedure performed
when the pathological substance is found in the circulation o The purpose is to remove the agent in the plasma, such as an
o The indication categories for therapeutic apheresis are as antibody, toxin, or abnormal protein, that is causing the
follows: clinical symptoms
o TPE is also used to replace a normal factor or substance that
Category I  Apheresis is standard and acceptable, either as may be missing or deficient in the patient’s plasma
primary therapy or as a first-line adjunct to other o During the TPE procedure, replacement fluids are being
initial therapies administered to maintain the patient’s intravascular volume,
 Efficacy is based on well-designed randomized resulting in dilution of plasma proteins
controlled clinical trials or a broad base of published Therapeutic plateletpheresis can be used to treat patients who
experience have abnormally elevated platelet counts with related symptoms
Category II  Apheresis is generally accepted in a supportive role or o During a plateletpheresis procedure, the platelet count will
as second-line therapy, rather than first-line therapy be decreased by 30% to 60%
Category III  Apheresis is not clearly indicated based on insufficient Therapeutic leukapheresis has been used to treat patients with
evidence, conflicting results, or inability to document a hyperleukocytosis, defined as a WBC or circulating blast count of over
favorable risk-to-benefit ratio 100,000/µL
 Decision-making should be individualized Erythrocytapheresis, or red cell exchange, removes a large number
Category IV  Apheresis has been demonstrated to lack efficacy or of RBCs from the patient and returns the patient’s plasma and
be harmful, and should be discouraged in these platelets along with compatible allogeneic donor RBCs
disorders o The procedure is most commonly performed in patients
 Clinical applications should be undertaken only under with sickle cell disease in order to decrease the number of
an approved research protocol hemoglobin S–containing RBCs, thereby treating or
preventing the complications (acute chest syndrome,

#MLSWATER2022 Page 7 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
impending stroke, unrelenting painful crisis) associated with Transfusion therapy is used primarily to treat two conditions:
the disease o Inadequate oxygen-carrying capacity because of anemia or
blood loss
The therapeutic goal is to decrease the level of hemoglobin S to less o Insufficient coagulation proteins or platelets to provide
than 30%: adequate hemostasis
 This is usually accomplished with a single red cell exchange Blood and blood products are considered to be drugs because of
procedure, requiring from six to ten RBC units, depending on the their use in the treatment of disease
patient’s age and red cell volume How do we prepare blood components product in transfusion
 The donor RBCs selected for transfusion should be ABO- and Rh- therapy?
compatible, relatively fresh (less than 10 days is preferable to
allow maximum in vivo survival), leukocyte-reduced, negative for Whole Blood
hemoglobin S (donor does not have sickle cell trait), and partially Whole blood should be used to replace the loss of both RBC mass
phenotype-matched for the Rh (C, c, E, e) and K1 antigens to and plasma volume
avoid future alloimmunization A definite contraindication to the use of whole blood is severe
 The procedure can be considered for treatment of overwhelming chronic anemia
malaria or Babesia infections For a 70-kg (155-lb) adult, each unit of whole blood should increase
o Both of these protozoa infect red blood cells, and a the hematocrit level 3% or hemoglobin 1 g/dL.
pronounced parasitemia can occur o After transfusion, the increase may not be apparent until 48
o Red cell exchange has been shown to be beneficial for to 72 hours when the patient’s blood volume adjusts to
treating these patients when the parasite load is greater normal
than 10%
o A 1.5- to 2-volume red cell exchange should significantly If given that the RBC mass is only affected, plasma volume is
decrease the parasite load increased already. Transfusing and inducing can’t be done to the
 Red cell exchange can be used to remove incompatible RBCs from whole blood of the patient
a patient’s circulation Patients suffering from severe chronic anemia has a reduce RBC
count/mass but even though the RBC mass is reduced but the plasma
volume is increased due to the compensation of the body
The patient will suffer from circulatory volume overload once
TRANSFUSION THERAPY transfused with plasma, causing for the patients to develop adverse
reaction such as pulmonary edema and heart failure
TRANSFUSION MEDICINE
It happens mostly in patients with pre-existing heart or kidney failure
Was created because there are people who are in need of blood and
other blood components in the treatment of disease
Red Blood Cells
A multi-disciplinary branch of medicine that is concern with the
RBCs are indicated for increasing the RBC mass in patients who
transfusion of blood and the blood components or products. Including require increased oxygen-carrying capacity
the proper selection and utilization of blood components as well as the The decreased RBC mass may be caused by:
removal of blood or blood components in the treatment of the o Decreased bone marrow production (leukemia or aplastic
diseases anemia)
o Decreased RBC survival (hemolytic anemia)
o Surgical or traumatic bleeding
#MLSWATER2022 Page 8 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Transfusion of RBCs is contraindicated in patients who are well Freezing RBCs allows the long-term storage of rare blood donor
compensated for the anemia units, autologous units, and units for special purposes, such as
RBCs should not be used to treat nutritional anemia, such as iron intrauterine transfusion
deficiency or pernicious anemia, unless the patient shows sign of
decompensation (need for increased oxygen-carrying capacity) Platelets and Plateletpheresis
RBC transfusion is not to be used to enhance general well-being, Platelets are essential for the formation of the primary hemostatic
promote wound healing, prevent infection, expand blood volume plug and maintenance of normal hemostasis
when oxygen-carrying capacity is adequate, or prevent future Patients with severe thrombocytopenia (low platelet count) or
anemia. abnormal platelet function may have petechiae, ecchymoses, and
Each unit of transfused RBCs is expected to increase the hemoglobin mucosal or spontaneous hemorrhage
level 1 g/dL and the hematocrit level 3% in the typical 70-kg (154-lb) The thrombocytopenia may be caused by decreased platelet
human, the same as whole blood production (e.g., after chemotherapy for malignancy) or increased
In pediatric patients, a dose of 10 to 15 mL/kg will increase the destruction (e.g., disseminated intravascular coagulation [DIC])
hemoglobin about 2 to 3 g/dL or the hematocrit 6% to 9% o Massive transfusion may also cause thrombocytopenia
because of the rapid consumption of platelets for hemostasis
Leukocyte-Reduced RBCs and the dilution of the platelets by resuscitation fluids and
Donor leukocytes may cause: RBC transfusion
o Febrile nonhemolytic transfusion reactions Platelet transfusions are indicated for patients who are bleeding
o Transfusion-associated graft-versus-host disease (TA-GVHD) because of thrombocytopenia or abnormally functioning platelets
o Transfusion-related immune suppression One plateletpheresis should increase the adult patient’s platelet
In addition, human leukocyte antigens (HLA) are responsible for HLA count to 20,000 to 60,000/µL
alloimmunization o Each unit of platelets from whole blood must contain at least
Leukocytes may harbor cytomegalovirus (CMV) 5.5 × 10¹⁰ platelets and should increase the platelet count by
o To reduce HLA alloimmunization and CMV transmission, the 5,000 to 10,000/µL in a 70-kg human
leukocyte content must be reduced to less than 5 × 10⁶,
which can be achieved by using one of several leukocyte Granulocyte Pheresis
reduction filters Newborn infants may develop overwhelming infection with
o With these filters, most RBC units are less than 1 × 10⁶; neutropenia because of their limited bone marrow reserve for
some are 1 × 10⁴ leukocytes neutrophil production
o In addition, neonatal neutrophils have impaired function
Washed RBCs and Frozen/Deglycerolized RBCs o Studies show granulocyte transfusions to be beneficial for
Patients who have severe allergic (anaphylactic) transfusion these patients
For an adult or a child, the usual dose is one granulocyte pheresis
reactions to ordinary units of RBCs may benefit from receiving
product daily for 4 or more days
washed RBCs
o For neonates, a portion of a granulocyte pheresis unit is usually
The washing process removes plasma proteins, the cause of most
given once or twice
allergic reactions
Granulocyte components should be administered as soon as
Washed RBCs are used for the rare patient who has had moderate
possible and within 24 hours of collection
to severe allergic transfusion reactions and has anti-IgA antibodies
The granulocyte pheresis needs to be crossmatched because of the
because of IgA deficiency
significant content of RBCs
#MLSWATER2022 Page 9 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
The neutrophil count will increase to 1,000/µL or more in response Fibrinogen replacement may be required in patients with liver
to infusion of granulocyte colony-stimulating factor (GCSF)– failure, DIC, or massive transfusion and in rare patients with
mobilized granulocyte pheresis congenital fibrinogen deficiency
Cryoprecipitate was used as a source for fibrin sealant, which uses
Plasma cryoprecipitate as the source of fibrinogen
Plasma includes fresh frozen plasma, plasma 24 (frozen within 24 Cryoprecipitate was originally prepared as a source of factor VIII
hours), and thawed plasma Each unit of cryoprecipitate must contain at least 80 units of factor
o Plasma and plasma 24 contain all coagulation factors VIII
o After fresh frozen plasma and plasma 24 are thawed, they can
become thawed plasma and stored for 5 days at 4°C Factor VIII
o The 5-day storage reduces outdating and allows rapid response Patients with hemophilia A or factor VIII deficiency have
to urgent orders for bleeding patients spontaneous hemorrhages that are treated with recombinant or
o Thawed plasma after 5-day storage has less factor V and VIII human plasma–derived factor VIII replacement
but is still therapeutic Factor VIII is treated by different methods, such as pasteurization,
Plasma can be used to treat multiple coagulation deficiencies nanofiltration, and solvent detergent, to ensure sterility for HIV,
occurring in patients with liver failure, DIC, vitamin K deficiency, hepatitis B virus, and hepatitis C virus (HCV)
warfarin overdose, or massive transfusion Both the plasma derived and recombinant factor VIII are stored at
Plasma is the product of choice for patients with multiple-factor refrigerator temperatures and are reconstituted with saline at the
deficiencies and hemorrhage or impending surgery time of infusion
Usually 4 to 6 units of plasma will effectively control hemostasis
Congenital coagulation factor deficiencies are rarely treated with Factor IX
plasma, because the dose requirement for surgical procedures and Factor IX complex (prothrombin complex) is prepared from pooled
serious bleeding is so great as to cause pulmonary edema as a result plasma using various methods of separation and viral inactivation
of volume overload, even in a young individual with a healthy The prothrombin complex contains factors II, VII, IX, and X;
cardiovascular system however, the product is recommended for factor IX–deficient
Plasma is sometimes used as a replacement fluid during plasma patients (hemophilia B), patients with factor VII or X deficiency
exchange (rare), and selected patients with factor VIII inhibitors or reversal of
Plasma should not be used for blood volume expansion or protein warfarin overdose
replacement because safer products are available for these
purposes—serum albumin, synthetic colloids, and balanced salt Antithrombin and Other Concentrates
solutions—none of which transmit disease or cause severe allergic Antithrombin is a protease inhibitor with activity toward thrombin
reactions or transfusion-associated acute lung injury Heparin accelerates the binding and inactivation of thrombin by
Plasma should be ABO-compatible with the recipient’s RBCs, but the antithrombin.
Rh type can be disregarded The hereditary deficiency of antithrombin is associated with venous
thromboses, whereas the acquired deficiency is seen most
Cryoprecipitate frequently with DIC
Cryoprecipitate is used primarily for fibrinogen replacement Thawed plasma is an alternative source of antithrombin
The AABB requires that at least 150 mg of fibrinogen be in each unit Protein C and protein S are vitamin K–dependent proteins
of cryoprecipitate synthesized in the liver

#MLSWATER2022 Page 10 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE
Protein S functions as a cofactor for activated protein C, which, in
turn, inactivates factors V and VIII, thus preventing thrombus
formation

Albumin
Albumin is prepared by chemical and physical fractionation of pooled
plasma
Albumin is available as a 5% or a 25% solution, of which 96% of the
protein content is albumin
Albumin may be used to treat patients requiring volume replacement
Albumin can also be used in the treatment of burn patients to replace
colloid pressure

Immune Globulin
Immune globulin prepared from pooled plasma is primarily IgG
Products are available for intramuscular or intravenous
administration
o The intramuscular product must not be given intravenously
because severe anaphylactic reactions may occur
o The intravenous product must be given slowly to lessen the risk
of reaction
Immune globulin is used for patients with congenital
hypogammaglobulinemia and for patients exposed to diseases such
as hepatitis A or measles
For hypogammaglobulinemia, monthly injections are usually given
because of the 22-day half-life of IgG
The recommended dose is 0.7 mL/kg intramuscularly or 100 mg/kg
intravenously
The intravenous preparation of immune globulin is used increasingly
in the therapy of autoimmune diseases, such as immune
thrombocytopenia and myasthenia gravis
Rh immune globulin (RhIg) was developed to protect the Rh-
negative female who is pregnant or who delivers a Rh-positive
infant
Rh immune globulin products, which can be administered
intravenously or intramuscularly, are approved for use in idiopathic
thrombocytopenic purpura patients who are Rh (+)

-END OF TRANSCRIPTION-

#MLSWATER2022 Page 11 of 11
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
MLS 18, April 6, 2021
BSMLS 3B | BY MLSWATER
University of San Agustin-Iloilo`

Legend:
Transcription Bullet During Transfusion
Notes/Module Packet Bullet Positive identification
 PPT  Identification of the patient and also the patient’s blood specimen
prior to transfusion as well as the blood unit that is used in transfusion
MODULE OUTLINE
I. Adverse Effects (page 4)
Clerical errors represent the main cause of transfusion related death
A. Immediate HTR
and acute hemolytic reaction. Final clerical check is performed at
B. Delayed HTR
II. Other Related Disease (page 5) the bedside patient done by nurse and it also double checks the
A. Transfusion-Transmitted Disease blood bag tag attached to the component to be transfused against
B. Hemolytic Disease of the Newborn the patient’s arm band
C. Autoimmune Hemolytic Anemia The medtech’s job is only in the issuance of the blood, to check if
the blood is viable (check for presence of any contaminants,
LEARNING OUTCOMES bacterial contamination and clots that might cause problem during
At the end of this module, the student shall be able to: transfusion).
1. Define transfusion reaction. The specimen must be collected within 3 days of the scheduled
2. Explain the risks of transfusions. transfusion and the blood can’t be used when it exceeds 3 days. The
3. Compare and contrast immediate and delayed HTR; HTR and non- donor’s blood must be kept stoppered or sealed and stored in 1-6°C
HTR
for at least 7 days post transfusion
4. Differentiate the clinical signs & symptoms of the various types of
Even though the transfusion is done, pre-transfusion blood
transfusion reactions.
5. List laboratory findings associated with acute transfusion reactions specimen is prohibited to put into waste
All specimen that is used in pre-transfusion must be kept for 7 days
because if ever there are adverse reactions during transfusion, there
WHAT TO DO DURING TRANSFUSION? would be specimen that can be used as the basis to compare the
after checking if the donated blood can be transfused to the recipient, result in the post transfusion specimens
proper procedure in the transfusing of the blood must also be done
aside from screening or testing the blood being qualified in transfusion, Venous access
proper procedure in transfusing the blood must be observed Should be established before a blood component is issued
take into consideration the precautionary measures transfusion, as well The selection of the location and the type of access are dependent
as cases where transfusion reaction might occur on the volume, timing expected duration of the transfusion therapy
Peripheral access with an 18 gauge needle or catheter is typically
sufficient

#MLSWATER2022 Page 1 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
It is desirable for high volume administration or for long term Automatic temperature control is set when the blood is warmed at
therapy 42°C
Never use water bath as blood warmer or microwave
Intravenous transfusion device
Should be set before the blood is issued
Rate and length of transfusion
Isotonic saline or 5% albumin should be used as an intravenous
Blood components are infused slowly for the first 10-15 minutes
solution to dilute the blood components. Because it is not
Approximately 2mL for the first 15mins.
preferable to transfuse blood that is too viscous since it is not
Slow transfusion is needed for the nurses and physician to observe
advisable
any sign of transfusion reaction
Do not use 5% dextrose solution in water also known as D5W (a
Subsequently the administration rate maybe increased and the
hypotonic solution)
desirable time to complete the red cell transfusion should be
o that can cause hemolysis of the RBC
finished within 2 hours
Do not use ringer solution, since it contains calcium which initiates
For platelets and plasma, since it is less in volume compared to red
coagulation
cell transfusion, the transfusion of platelets and plasma should fall
No medication must be added or administered in the same line with
within 30-60 minutes
the blood component
NOTE: Any transfusion should be completed within 4 hours of
initiation
Clot-screen filters o Exceeding to 4 hours would put the patients at risk for
All blood components must be filtered to remove the clots by using bacteremia or septicemia
the clot screen filters. At least 75% of the transfused red cells remains in the recipient’s
70 -170 microns of filters are used to remove gross clots and cellular circulation 24 hours after the transfusion
debris
SPECIAL CONSIDERATION IN TRANSFUSION
Vital signs Emergency transfusion
Pulse rate, respiratory rate, blood pressure, temperature are If patients Abo and Rh group is unknown, during emergency
checked by the nurse to note if there would be any signs of transfusion patients are given group O Rh negative red cells since
transfusion reaction group O/ blood type O individuals are considered to be as universal
donor (they do not contain any antigen)
Blood warmers Emergency situations or transfusion require group O Rh negative
Cold blood can cause hypothermia in the patient red cell if the patient’s Abo or Rh is unknown
Hypothermia can lead to cardiac arrhythmia and haemorrhage Massive transfusion
Blood should be set at 37°C defined as the administration of enough blood or components
Blood must not be allowed to warm above 38°C because it can within less than 24 hours to reconstitute a complete volume
induce physical or chemical damage in our cells, that can make the replacement
blood unit not viable to transfusion anymore in adults, this requires 8-10 units of blood

#MLSWATER2022 Page 2 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
Adverse Effects of Massive Transfusion Intrauterine Transfusion
1. Citrate toxicity and Hypocalcemia done when the baby inside the womb is suffering severe anemia
o Citrate toxicity associated with hemolytic disease of the fetus or the newborn
caused by the presence of citrate in the anticoagulant Is done by:
preservative solution o intraperitoneally inject RBCs into the fetal peritoneal cavity
patients experiencing hypothermia, 2,3-DPG depletion, where RBC can be absorbed to the circulation or perform
coagulation factor depletion, as well as depletion of platelets cordocentesis
due to delusional effects because of extremely massive  Process where donor RBC is directly injected into
transfusion the fetal umbilical vein
there could also be accumulation of biochemical and
microaggregates Purpose of the Intrauterine Transfusion
to maintain fetal hemoglobin above 10g/dL
Neonatal RBC transfusion once initiated, the procedure must be repeated every 2-4 weeks
Done in neonates less than seven days old to reduce the risk of until the 34-36 gestation week is reached or until the fetal lung has
hyperkalemia and to maximize 2,3-DPG matured
For O-negative or blood types compatible with the mother, the
infant is given CMV-negative or leukocyte-reduced blood WHAT TO DO WHEN THIN GS GO WRONG?
o should be hemoglobin S negative for hypoxic and acidotic
newborns Bedside Procedures Transfusion Reactions
Prepare aliquots since neonates require small volume of RBCs Investigation Work up
o dosage: 10-15 mL / kg over 2-3 hours 1. Stop transfusion 1. Clerical checks
2. Keep intravenous line open 2. Visual inspection
Exchange Transfusion
with physiologic saline 3. DAT
used in the treatment for both anemia and hyperbilirubinemia that
4. Repeat ABO /Rh
characterizes hemolytic disease of the fetus and the newborn 3. Notify patient’s physician and
5. Repeat compatibility testing
small amounts of blood are removed from the baby and then blood bank 6. Repeat crossmatch
exchanged with the donor’s blood 4. Take care of the patient per 7. Bilirubin test in blood
Main purpose or the indication: physician’s order 8. Urine test
o to replace the antibody-coated red cells with the compatible 5. Perform bedside clerical checks 9. Hemoglobin and hematocrit
donor cells 6. Specimen to be submitted Post
o to remove bilirubin in order to prevent Kernicterus transfusion venous blood. First
o to remove the circulating maternal antibody in the baby’s
voided urine and blood bags
plasma
7. Document reaction
o to suppress the erythropoiesis or the production of
incompatible red blood cells
Greater than 0.5 mg/dL per hour rise in bilirubin or 10 mg/dL in the
first 24hrs amount of bilirubin would be indicative that the baby is
in need of exchange transfusion

#MLSWATER2022 Page 3 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
ADVERSE EFFECTS Transfusion-Related Acute Lung Injury (TRALI)
Transfusion Reaction attributed to the administration of donor plasma containing high
Defined as any transfusion-related adverse event that occurs during concentrations of leukoagglutinins
or after the transfusion of whole blood, blood components or Cause: anti-leukocyte antibodies act on endothelial cells of lungs;
human-derived plasma products. emboli form
They are classified according to the time interval between Remedy: Leukopoor RBCs
transfusion and the presentation of adverse effects
Non-Immunologic Reaction
IMMEDIATE (Acute) – DELAYED Bacterial Contamination
signs and symptoms presenting after 24 hours of transfusion Contamination of blood products from donors with transient
within 24 hours of transfusion. bacteremia during phlebotomy, preparation and processing,
Immunologic Non- Immunologic Non-Immunologic thawing.
Immunologic Cause: endotoxins of psychrophilic organisms; Yersinia
Hemolytic- Bacterial Hemolytic TA- Hemosiderosis enterocolitica, E. coli, Pseudomonas spp.
Febrile contamination TA-GVHD Disease Transmission Prevention: Aseptic techniques, visual inspection and infusion
within 4 hours
Allergic Circulatory Post-
Overload transfusion Transfusion Associated Circulatory Overload (TACO)
TRALI Purpura Associated with rapid infusion of large volumes of blood products.
PCITR Children, elderly patients and cardiac disease patients
Treatment can include intravenous diuretics and therapeutic
Immediate (Acute) Transfusion Reaction phlebotomy
Immunologic Reaction
Febrile Non-Hemolytic Physical or Chemical Induced Transfusion Reaction (PCITR)
Increase in temperature of 1 degree Celsius associated with Causes: mechanical damage (infusion through small bore), osmotic
transfusion that cannot be explained by other condition. or chemical damage, thermal trauma, citrate toxicity
Most common adverse reaction seen
Cause: Anti-leukocyte antibodies Includes physical damage to the RBC due to intravascular lysis
Remedy: Leukopoor RBCs Cause: proliferation of T-cells reacts against foreign tissue of the
host recipient
ALLERGIC VS. ANAPHYLACTIC REACTIONS Prevention: Irradiated blood components
ALLERGIC ALLERGIC
Reaction between recipient Afebrile reaction that occurs only Delayed Immunologic Reaction
antibody and transfused donor after infusion of only few mL of Immunologic Reaction
plasma proteins blood Transfusion-Associated Graft vs. Host Disease (TA-GVHD)
Cause: Donor plasma with foreign Cause: IgA deficient patient with Certain susceptible recipients with compromised immune systems
proteins anti-IgA are transfused with blood or blood components containing
Remedy: Washed RBCs Remedy: Washed RBCs
#MLSWATER2022 Page 4 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
immunocompetent lymphocytes which engraft in recipient’s tissue Hepatitis
and multiply. Inflammation of the liver
Cause: Proliferation of T cells that reacts against foreign tissue of Symptoms: jaundice, dark urine, hepatomegaly, anorexia, malaise,
the host recipient fever, etc.
Prevention: Irradiated blood components
Hepatitis A
Picornaviridae
Post- Transfusion Purpura Small, non-enveloped, single stranded enterovirus RNA
Previously immunized patients to platelet antigens through Most common
pregnancy or transfusion produce antibodies once stimulated, Not screened for blood units
destroying patient’s own platelets.
Cause: Platelet antibodies
Hepatitis B
Therapy: Exchange transfusion, corticosteroids, intravenous
A partially double stranded circular DNA virus of the Hepadnaviridae
immunoglobulin

Hepatitis C
Non-Immunologic Reaction
Flaviviridae virus family. RNA virus.
TA- Hemosiderosis Diagnosis depends on biochemical changes suggestive of HCV,
Iron overload accumulating in the mitochondria of cells in organs detection of HCV RNA or anti-HCV in serum.
like the liver, heart and endocrine gland. EIA, ChLIA and RIBA (Recombinant immunoblot assays)
Individuals that are chronically transfused are at risk example
thalassemia major, sickle-cell anemia and hemoglobinopathies. Hepatitis D, E, G
Iron chelating agent like Desferrioxamine can be used for therapy. HDV is a defective, single-stranded RNA virus that is found only in
the patients with HBV infection.
OTHER RELATED DISEASE o It requires HBsAg in order to synthesize an envelope
Transfusion-Transmitted Disease protein. It was previously called the delta antigen. If HBV
Hepatitis B,C,D and HDV are contracted concurrently, this co-infection.
CMV HEV is a member of the Calciviridae family of nonenveloped RNA
EBV viruses.
HTLV 1 and 2 GB virus C (GBV-C) and hepatitis G virus (HGV) are two genotypes of
HIV the same enveloped RNA virus that belongs to the Flaviviridae
T. pallidum family.
Plasmodium spp.
B. microti
T. cruzi
T. gondii

#MLSWATER2022 Page 5 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
o Primary infection with HIV may be asymptomatic or result in a
Hepatitis Route of Transmission mild, chronic lymphadenopathy with symptoms similar to those
seen in infectious mononucleosis.
A fecal/oral  Symptoms may occur within 6 to 12 weeks of infection
and persist for a few days to 2 weeks. HIV enters the cell
B parenterally
by the binding of the virus glycoprotein 120 with cell
surface receptors. Cells possessing these receptors
C parenterally
include CD4+ lymphocytes, macrophages, and other
D parenterally antigen-presenting cells.
 Expansion of donor screening protocols in 1996 to include
E fecal/oral p24 core antigen testing reduced the window period from
an estimated 22 days to 16 days
G parenterally
Refer to Appendices for Decision Tree for Anti-HIV-1/HIV-2 testing of donor
blood.
Diagnosis
HBV DNA first marker to appear- PCR
HBsAg- 2 to 12 weeks post-exposure during acute stage; HTLV I and II (Human T-Cell Lymphotropic Viruses Types I/II)
undetectable in 12 -20 weeks after the development of anti-HBsAg HTLV-I and HTLV-II are RNA retroviruses.
HbeAg appears after the HBsAg in recovering patients, disappears o HTLV-I causes a T-cell proliferation with persistent infection.
before HbsAg. Once the RNA has been transcribed into DNA, it is integrated
HbcAg is present in the serum but is undetectable. randomly into the host cell’s genome.
Once integrated into the DNA, the provirus can either complete its
HIV Type 1 and 2 replication cycle or remain latent for many years.
HTLV-I was the first retrovirus to be associated with a human
HIV is a retrovirus that is spherical in shape, with an approximate
disease.
diameter of 100 nm. That association was with adult T-cell lymphoma/ leukemia (ATL), a
It consists of an envelope of glycoproteins, core proteins, and an highly aggressive, mature T-cell non-Hodgkin’s lymphoma with a
inner core of viral RNA and reverse transcriptase. leukemic phase.
o Infection with the virus causes a slowly progressing immune HTLV-II is similar to type-I and is prevalent in intravenous drug
disorder. users.
o The causative viruses, HIV-1 and HIV-2, are similar in structure,
Refer to Appendices for Flowchart of HTLV Testing
varying primarily in the envelope proteins
o The use of very sensitive serologic testing in screening the blood
supply has resulted in an extremely low risk of HIV transmission
o Positive screening tests are repeated in duplicate, and if at least
one of the duplicates also tests positive, WB or
immunofluorescent antibody assay is used for confirmation.
#MLSWATER2022 Page 6 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
West Nile Virus EBV
WNV is a member of the Flavivirus family and is a human, avian, and EBV has been called the “kissing disease” because the virus usually
equine neuropathogen. replicates in the cells of the oropharynx, possibly in infected B cells.
It is a single-stranded RNA lipid-enveloped virion that is common in The virus is shed in the saliva and is most frequently associated with
Africa, West Asia, and the Middle East. infectious mononucleosis.
WNV is usually subclinical but may cause a mild flu-like disease.
Mosquitoes have been found to carry WNV, the genus Culex is the Human Herpesvirus 6 (HHV-6) and 8 (HHV-8) Human herpesvirus 6
chief vector. (HHV-6)
The infection in humans has an incubation period of approximately
Is a very common virus that causes a lifelong infection.
3 to 14 days following the mosquito bite, with symptoms lasting 3 to
After infection, the virus replicates in the salivary gland and then
6 days.
remains latent in lymphocytes, monocytes, and perhaps other
o Other animals can become infected, including horses, cats,
tissues, without obvious pathology.
dogs, bats, chipmunks, skunks, squirrels, and domestic birds
HHV-6 causes roseola infantum, also known as exanthem subitum
and rabbits.
or sixth disease.
CMV
Herpesvirus group Syphilis
CMV can remain latent in the tissues and leukocytes for years, with
Treponema pallidum, the causative agent of syphilis, is a spirochete.
reactivation occurring due to severe immune system impairment.
It is usually spread through sexual contact but can be transmitted
Those at the highest risk of a CMV infection are individuals receiving
through blood transfusions.
allogeneic marrow transplants and the fetus.
Spirochete cannot live in blood stored for 72 -96 hours at 1-6⁰C-
o CMV-seronegative recipients transplanted with
which would make platelets the only component capable of
CMVseronegative allogeneic marrow are at risk if they
transmitting the infection.
receive untested and non-WBC-reduced blood components.
Still part of the test since donor positive for syphilis are considered
high risk individuals.
Parvovirus B19
Human B19 parvovirus (B19) is a small, single-stranded DNA non- Tick-Borne Bacterial Agents
enveloped virus and is the only known pathogenic human
Lyme disease, Rocky Mountain spotted fever (RMSF), and
parvovirus.
ehrlichioses are all bacterial diseases spread by a tick bite.
It causes a common childhood illness called “fifth disease” and is
Lyme disease is caused by the spirochete Borrelia burgdorferi and
usually transmitted through respiratory secretions.
RMSF (Rickettsia rickettsii) and ehrlichioses (Ehrlichia species) are
o Fifth disease presents with a mild rash described as “slapped
caused by bacteria that are obligate intracellular pathogens.
cheek” when occurring on the face and a lacy red rash when
occurring on the trunk and limbs.
o The virus must enter the cell through a specific cell receptor. B19 Transfusion-Associated Parasites
parvovirus enters the red blood cell (RBC) via the P antigen and At least three parasites have been associated with transfusionassociated
replicates in the erythroid progenitor cells. infections:
1. Babesia microti
Babesiosis, a zoonotic disease, is usually transmitted by the bite of
an infected deer tick.
#MLSWATER2022 Page 7 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
Infection is caused by the protozoan parasite, Babesia, which infects During gestation, and particularly at delivery when the placenta
the RBCs. separates from the uterus, variable numbers of fetal RBCs enter the
Babesia infection may also be acquired by blood transfusion and maternal circulation
solid organ transplant. These fetal cells, carrying D antigen inherited from the father,
immunize the mother and stimulate the production of anti-D.
2. Trypanosoma cruzi Once the mother is immunized to D antigen, all subsequent
Trypanosoma cruzi is a flagellate protozoan that is the etiologic offspring inheriting the D antigen will be affected.
agent of Chagas disease (American trypanosomiasis). The maternal anti-D crosses the placenta and binds to the fetal Rh-
The reduviid bug bite produces a localized nodule, referred to as a positive cells.
chagoma. The sensitized RBCs are destroyed by the fetal reticuloendothelial
system, resulting in anemia
3. Malaria (Plasmodium species)
Malaria, another intraerythrocytic protozoan infection, may be
caused by several species of the genus Plasmodium (P. malaria, P.
falciparum, P. vivax, and P. ovale).
Natural transmission occurs through the bite of a female Anopheles
mosquito, but infection may also occur following transfusion of
infected blood.

Prion Disease
Creutzfeldt-Jakob Disease (CJD)
Creutzfeldt-Jakob Disease (CJD) is one of the transmissible
spongiform encephalopathies (TSE). These are rare diseases
characterized by fatal neuro-degeneration resulting in spongelike
lesions in the brain.

Hemolytic Disease of the Newborn and Fetus (HDFN)


 destruction of the RBCS of the fetus and neonate by the antibodies
produce by the mother
 Only the antibodies if the immunoglobulin G (IgG) class are actively
transported across the placenta

Usually in Rh(D) HDN, the Rh-positive firstborn infant of a Rh-


negative mother is unaffected because the mother has not yet been
immunized.

#MLSWATER2022 Page 8 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
Characteristics of HDN INDICATION
Anemia Antenatal RhIg should be given early in the third trimester, or at
• compensated anemia (increased immature RBCs leads to about 28 weeks’ gestation. The dose does not pose a risk
erythroblastosis fetalis) to the fetus, inasmuch as this amount will cause a titer of
Generalized edema and cardiac feature (hydrops fetalis) only 1 or 2 in the mother.
Increased urinary bilirubin Post-partum The Rh-negative nonimmunized mother should receive
Deposition in brain tissue ( kernicterus) RhIg soon after delivery of an Rh-positive infant. The
Hepatosplenomegaly (extramedullary hematopoiesis) recommended interval is within 72 hours after delivery.

Serological Testing Dose and Administration


Mother Newborn Infant 15 mL of packed RBCs or 30 mL of whole blood
 ABO, Rh, Antibody Screen  ABO This is equal to 300 g of the World Health Organization (WHO)
 Antibody Identification  Rh reference material.
 Paternal Phenotype and  DAT
Genotype The total fetal blood volume is estimated to be less than 5 mL at 12
 Elution weeks.
 Fetal DNA Testing
 Antibody Titers o The microdose can be used for abortions and ectopic
pregnancies before the 12th week of gestation.
RhIg An IV preparation of RhIg is approved for use in the United States.
Active immunization induced by RBC antigen can be prevented by This product also contains 300ug in each vial and can be
the concurrent administration of the corresponding RBC antibody. administered either intramuscularly or intravenously.
This principle has been used to prevent immunization to D antigen Massive fetomaternal hemorrhages of more than 30 mL of whole
by the use of high-titered RhIg. blood occur in less than 1 percent of deliveries.
These massive hemorrhages can lead to immunization if adequate
Mechanism RhIg is not administered.
o A maternal sample should be obtained within 1 hour of
o The administered RhIg attaches to the fetal Rh-positive RBCs in delivery to test for massive fetomaternal hemorrhage by a
the maternal circulation. The antibody coated RBCs are removed
screening test, such as the rosette technique. If positive,
by the macrophages in the maternal spleen. The RBC antigens
quantitation of the hemorrhage must be done by Kleihauer-
are thus unavailable for dendritic cells to present antigen to T
helper cells. Betke or similar test or by flow cytometry

Kleihauer-Betke test
Kleihauer-Betke test, a maternal blood smear is treated with acid and then
stained with counterstain.
Fetal cells contain fetal hemoglobin (Hgb F) that is resistant to acid
and will remain pink.

#MLSWATER2022 Page 9 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
The maternal cells will appear as ghosts. Bilirubin at
Normal Elevated
birth
Anemia at
No Yes
birth
DAT Weakly positive/ negative Positive
Spherocytosis Yes Rare
Phototherapy Phototherapy,
Therapy Exchange/ intrauterine
transfusion

Immune Hemolytic Anemia


A simpler way to calculate the dose is to multiply the fetal cell
Shortened RBC survival mediated through immune response.
percentage by 50, which gives the volume of fetomaternal
specifically by humoral antibody
hemorrhage in milliliters.
If needed, additional vials of RhIg should be administered within 72 3 TYPES
hours of delivery or as soon as possible. Alloimmune Patient produces alloantibodies to foreign or non-self
The RhIg must be injected according to the product label. The IV RBC antigens introduced into the circulation, most often
product can also be given intramuscularly. through transfusion or pregnancy
The intramuscular form must be given intramuscularly only.
IV injections of intramuscular preparations can cause severe
anaphylactic reactions because of the anticomplementary activity of Autoimmune Patient produces antibodies against his or her own RBC
these products. antigens
RhIg also contains IgA and may be contraindicated in patients with
anti-IgA and IgA deficiency who have had anaphylactic reactions to Drug-induced production of antibodies to a particular drug or drug
blood products. complex with ensuing damage to the individual’s RBCs

Comparison of ABO vs. Rh HDN Refer to Appendices for Diagram on Drug-Induced Hemolytic Anemia

Characteristics ABO Rh Drug-Dependent or Immune Complex (“Innocent Bystander”)


Non immune/immune IgG Immune IgG anti-D Mechanism
Antibody
anti-A, B The antibody (IgG or IgM) produced recognizes determinants on the
Mother= O Mother= Rh negative drug. If the patient ingests the same drug (or a drug bearing the same
Blood group haptenic group) after immunization, the formation of a drug-antidrug
Baby= A/B/AB Baby= Rh positive
First pregnancy and First pregnancy not complex may occur.
Obstetric The complement cascade may be activated because of this antigen-
subsequent pregnancies affected; rare
history antibody interaction. RBCs are thought to be involved in this process
may be affected
Disease only as “innocent bystanders.”
predicted by No Yes
titers

#MLSWATER2022 Page 10 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
The soluble drug-antidrug complex nonspecifically adsorbs loosely to
the RBC surface. Complement, when activated, sensitizes the cell and
may cause lysis.

Membrane Modification (Nonimmunologic Protein Adsorption)


It is hypothesized that the cephalosporins, especially cephalothin
(Keflin), both operate through the drug-adsorption mechanism and are
able to modify RBCs so that plasma proteins (e.g., IgG, IgM, IgA, and
complement) can bind to the membrane.

Autoimmune Hemolytic Anemia (AIHA)


Warm Autoimmune Hemolytic Anemia (WAIHA)
DAT: Positive
Donath- Landsteiner test: Negative
IgG antibody
Optimal temperature: 37 degrees Celsius
Rh, Kell
extravascular hemolysis; splenic

Cold Hemaglutinin Disease


DAT: Positive
Donath- Landsteiner test: Negative
IgM antibody
Optimal Temperature: 0-4 degrees Celsius
I blood group
Extravascular hemolysis; hepatic

Paroxysmal Cold Hemoglobinuria


DAT: Positive
Donath- Landsteiner test: Positive
Involves biphasic IgG
May occur on both 0-4 degrees Celsius; 37 degrees Celsius
P blood group
Intravascular

#MLSWATER2022 Page 11 of 12
MODULE 5: BLOOD DONATION & TRANSFUSION PRACTICE (part 4)
APPENDICES

Softcopy version:
https://drive.google.com/drive/folders/12nb8DHOKeTirLYYsWSum79-
QBcUZC-oe?usp=sharing

-END OF TRANSCRIPTION-

#MLSWATER2022 Page 12 of 12

You might also like