Blood System
Blood System
Blood System
Embryology
Sites of Hematopoiesis:
In the embryo Hemopoiesis occurs at different stages in the yolk sac, the
liver, the spleen, lymph nodes and the bone marrow.
Erythrocytes,
granulocytes, bone marrow
In the adult monocytes and platelets
In the lymph nodes, spleen, thymus
the lymphocytes and lymphatic nodules of the
gastrointestinal tract.
Embryo hematopoiesis:
1) In developing embryos, blood formation occurs in aggregates of blood cells in
the yolk sac, called blood islands.
2) As development progresses, blood formation occurs in the spleen, liver, and
lymph nodes.
3) When bone marrow develops, it eventually assumes the task of forming most
of the blood cells for the entire organism.
Blood cell development begins as early as the seventh day of embryonic life.
Red blood cells are essential in delivering oxygen to tissues and the
development of vascular channels during embryogenesis.
First waves:
Conversion of primitive haematopoiesis into definitive haematopoiesis.
The first wave of primitive hematopoietic and endothelial cell development occurs
via signals to the extraembryonic, endodermal yolk sac within the first two weeks
of gestation, which results primarily in the formation of:
• Primitive erythroid cells (Eryp).
• Megakaryocytes.
• Macrophages.
• Endothelium.
These EryP cells are distinct from their erythroid progenitors in that they are:
1. Larger.
2. Nucleated.
3. Have embryonic globins.
4. Are detected only in the yolk sac.
EryP help in the formation of structures called blood islands in which the
centrally placed cells give rise to erythroid and myeloid cells while peripherally
placed cells form endothelial cells that form these channels.
These blood islands fuse to form vascular channels that span throughout the
yolk sac.
Second wave
The second wave of definitive hematopoiesis replaces primitive hematopoiesis
and the first wave of definitive hematopoiesis.
Hematopoietic stem cells (HSC) emerge from a specialized hemogenic
endothelium within a limited region of the developing aorta's ventral wall called
the para-aortic splanchnopleure.
• The aorta-gonad-mesonephros (AGM) region develops from the para-aortic
splanchnopleure and produces HSC.
These cells colonize the fetal liver by the 6th or 7th week of gestation, where
they cycle at a continuous pace and begin to differentiate.
At this point, the liver becomes a significant source of hematopoietic stem
cell production.
HSC also colonize the spleen around 10-28 weeks or 20th week and produce
red cells for a brief period.
A vital organ that HSC starts colonizing around this time is the bone marrow.
HSC seeding ( )تنبتin the marrow is critical because it is the bone marrow that will
predominate in erythropoiesis as gestation advances ()تقدم.
The fetal liver provides the microenvironment needed for expansion and
differentiation of definitive HSCs, from which definitive erythroid cells will
differentiate.
HSC in the fetal liver and spleen produces enucleated erythrocytes (EryD) that
rapidly outnumber ( )تفوق عددEryP cells in circulation.
Embryology
IN WEEKS 6-9
Local dilatations of the lymphatic channels form 6 primary lymph sacs:
Two jugular lymph sacs Pair Near the junction of the subclavian veins
with the anterior cardinals (future internal
jugular vein).
Two iliac lymph sacs Pair near the junction of the iliac veins with the
posterior cardinal veins
One retroperitoneal Single in the root of the mesentery on the posterior
abdominal wall
One cisterna chyli Single Dorsal to the retroperitoneal lymph sac, at
the level of the adrenal glands.
LYMPH VESSELS:
Grow out from the lymph sacs, along the major
veins to:
head, neck, and arms jugular sacs
lower trunk and legs iliac sacs
gut retroperitoneal and
cisternal sacs
The cisterna chyli is connected to the jugular lymph sacs by 2 large channels,
the right and left thoracic ducts.
Both the right and left thoracic ducts join the venous system at the angle of
the subclavian and internal jugular veins at the base of the neck
Duct Left lymphatic (thoracic) duct Right lymphatic duct
1. The caudal portion of the right thoracic duct.
Formed by cranial part of the right
2. The anastomosis.
thoracic duct
3. The cranial portion of the left thoracic duct.
Cisterna chyli; is the only sac that stay as it is, and doesn’t develop into other
new form.
Cisterna chyli drains lymph from all body parts except right upper
quadrent.
THE SPLEEN development:
Develops from an aggregation of mesenchymal cells in the dorsal mesentery of
the stomach at (15-17) weeks.
THE TONSILS
Palatine Tonsils (≈ 𝟏𝟒 𝒘𝒆𝒆𝒌𝒔). Develop from the second pair of
pharyngeal pouches.
The Tubal (Pharyngo Tympanic) Develop from aggregations of lymph
Tonsils. nodules around the openings of the
auditory tubes.
The Pharyngeal Tonsils (Adenoids). Develop from an aggregation of lymph
nodules in the nasopharyngeal wall.
The Lingual Tonsils. Develop from aggregations of lymph
nodules in the root of the tongue.
LYMPH NODULES also are seen in the mucosa of the digestive tract and
respiratory tract.
At about the end of week 5 The lymphatic system.
At about the end of week 3 CVS
about WEEKS (6-9) Form 6 primary lymph sacs.
At about month 3. sacs to lymph nodes
At about (15-17) weeks Spleen
at about 14 weeks Palatine Tonsils
Lymphatic system
Immunity or Resistance
Lymphatic capillaries
Made of a single layer of squamous epithelial cells
Slightly larger than blood capillaries
Cells overlap and act as one-way valves
Opened by pressure of interstitial fluid
Anchoring filaments attach cells to surrounding tissue
Channels of Lymphatics:
Lymphatics ultimately deliver lymph into 2 main channels:
Right lymphatic duct Thoracic duct (left)
Drains right side of head & neck, right Drains the rest of the body
arm, right thorax
Empties into the right subclavian vein Empties into the left subclavian vein
Cisterna chyli, it is the main duct for the return of lymph to blood.
Formation of lymph:
No capsule present
Diffuse lymphatic tissue Found in connective tissue of almost all organs.
Located where they come in direct contact with
antigens.
No capsule present
Lymphatic nodules Oval-shaped masses
Found singly or in clusters
Lymphatic organs Capsule present
Lymph nodes, spleen, thymus
Lymph Node:
Consists of connective tissue framework & numerous lymphocytes.
Bean shaped structures placed in pathway of lymphatic
vessels.
Enclosed by a fibrous capsule.
Cortex = outer portion
• Germinal centers produce lymphocytes
Medulla = inner portion
• Medullary cords
Lymph enters nodes through afferent lymph vessels, flows through sinuses,
exits through efferent lymph vessel.
Spleen:
Largest lymphoid organ
Encapsulated
Located between the stomach & diaphragm In upper left
quadrant of abdomen.
Structure is similar to a node, But no afferent vessels or
sinuses
Supporting Elements:
Capsule
Trabeculae
Trabecular network
Lymphocytes, macrophages, blood cells.
White pulp: Lymphoid Nodule (Malpighian corpuscle):
1. Germinal center
2. Central artery
Lymphatic tissue (lymphocytes and macrophages) little
islands, mostly B cells.
Functions of Spleen
Filtration of blood.
Immune response against antigens circulating in blood.
Site for production of B & T lymphocytes.
Formation of all blood cells during fetal life.
Only lymphocytes and monocytes after birth
Storage of blood {platelets} (~30%).
Can contain over one pint ( )مترof blood
Site of destruction of aged erythrocytes.
Thymus
Located behind the sternum in the mediastinum.
Development:
Infant conspicuous
Puberty maximum size
Maturity decreases in size
Thymic Lobule:
Lobules supporting stroma made by
epithelioreticular cells
• Cortex: densely packed small
lymphocytes.
• Medulla: Lymphocytes are less densely
packed.
Presence of Hassall’s corpuscles.
groups of epithelial cells within the thymic medulla.
Functions of thymus
• Provides the environment for stem cells where they can divide and mature
into T lymphocytes.
• Thymopoietin induces T cell production & maturation.
• Thymosin supports T cell activities.
Tonsils
– Multiple groups of large lymphatic nodules
Palatine tonsils Posterior-lateral wall of the oropharynx
Pharyngeal tonsil Posterior wall of nasopharynx
Lingual tonsils Base of tongue
Palatine Tonsil:
• Aggregation of lymphatic nodules within
diffuse lymphoid tissue.
• Covered by stratified squamous
epithelium.
• Tonsillar crypts (opening of numerous
mucous glands).
Functions of tonsil
Production of lymphocytes.
Immunological response against antigens & organisms coming in contact
with epithelium.
Physiology, biochemistry and histology
The blood, lymphatic, and immune systems have separate but interrelated
functions in maintaining a healthy environment within the body (homeostasis).
Blood is responsible for transporting oxygen (O2) and carbon dioxide
(CO2) and provides cells that defend against disease.
The lymphatic system is responsible for cellular communication by
delivering nutrients, hormones, and other needed products to body cells
while removing their waste products as it drains tissue fluid back to the
vascular system. It also provides the cells of the immune system needed to
defend the body against disease.
The immune system defends the body against disease. In its most simple form,
it uses barriers that exclude unwanted substances from entering the body.
In its most complex form, it uses cells of the lymphatic system to undertake
the complex processes that identify and destroy pathogens and protect the
body against future encounters by these same pathogens.
1) Blood
Blood is connective tissue composed of a liquid medium
called plasma in which solid components are suspended.
Blood is divided into two main components:
Although the size of white blood cells is greater than the size of red
blood cells, the density of red blood cells is greater than that of
hemoglobin (each red cell contains about 0.25 million hemoglobin molecules).
The solid components of blood include the following:
1) Red blood cells (erythrocytes)
2) Blood platelets (Thrombocytes)
3) White blood cells (leukocytes)
Granular leukocytes:
agranular leukocytes:
Medical notes:
1) The blood components are separated by a centrifuge.
2) The tube in which the sample is placed must contain anticoagulant materials
(practical example: EDTA tubes).
EDTA tubes contain substances that bind strongly with calcium (it helps in
coagulation greatly) and pull it out so that the sample does not coagulate.
Compartment Volume of Fluid Fluid Percentage Percentage of Body
(in Liters) of Body Weight
Total Body Fluid 42 100 60
Intracellular Fluid (ICF) 28 67 40
Extracellular Fluid (ECF) 14 33 20
Plasma 3.5 8.3 (25% Of ECF) 5
Interstitial fluid 10.5 25 (75% of ECF) 15
1) Blood volume:
Blood is thicker (more viscous) than water and flows more slowly than water.
Plasma at 37°C is about 1.8-times more viscous than water; therefore, the
relative viscosity of plasma (compared to water) is about 1.8
Whole blood viscosity (relative to water) = 4.5-5.5
Increased blood viscosity increases the risk of atherosclerosis
3) pH:
4) Color:
6) Osmotic pressure:
Composition of blood:
Cells or cells RBCs (4.8 – 6.5 million) 99%
fragments 45 % WBCs (4 – 11 thousands) < 1%
Platelets (150,000 – 450,000)
Water 91.5 %
Plasma protein 7%
Other solutes include:
Matrix (plasma) 55 %
• Electrolytes
• Organic nutrients and wastes 1.5 %
• Respiratory gases
• Vitamins
Note that:
Polycythemia = erythrocytosis
Anemia = erythropenia
Plasma proteins:
Plasma proteins: are proteins present in blood plasma.
1. Albumin (58%)
Albumin is the most abundant protein in plasma.
2. Globulins (37%).
𝜶 −Globulins Transport proteins
𝜷 −Globulins Coagulation factors
𝜸 −Globulins Defensive proteins = Immunoglobulins = Antibodies
3. Fibrinogen (4%).
4. Regulatory proteins (<1%)
C) LM appearance in smears:
Small basophilic fragments, often appearing in clusters.
hyalomere Lightly stained peripheral zone.
granulomere Darker-staining central zone rich in granules.
The shapes of RBCs can change remarkably as the cells squeeze through capillaries.
Does not have: nucleus, mitochondria, ribosomes, endoplasmic reticulum or Golgi
apparatus.
Persons living at high altitudes have greater numbers of RBCs,
Because the normal cell has a great excess of cell membrane for the quantity
of material inside, deformation does not stretch the membrane greatly and,
consequently, does not rupture the cell.
The shape is maintained by a cytoskeletal complex inside the plasma
membrane (involving spectrin, actin and other components).
Spectrin, actin problems cause anemia and spherocytosis.
The erythrocyte plasmalemma is consists of about 40% lipid, 10%
carbohydrate, and 50% protein.
Most of the proteins are integral membrane proteins, including: Ion
channels, the anion transporter called band 3 protein, and glycophorin A.
band 3 protein ABO incompatibility
glycophorin A Rh problems
4) LM appearance in smear:
Pink circle with light center (center is thinner
because of the biconcave shape) with NO
NUCLEUS.
5. TEM appearance:
Solid dark gray cytoplasm, because of highly
concentrated hemoglobin.
Genesis of Blood Cells (hematopoiesis):
In the early weeks of embryonic Primitive nucleated RBCs are produced in the yolk
life: sac.
During the middle trimester of The liver is the main organ for RBC production but
gestation: reasonable numbers are also produced in the spleen
and lymph nodes.
during the last month or so of RBCs are produced exclusively in the bone marrow.
gestation and after birth
The blood cells begin their lives in the bone marrow from a single type of cell called the
multipotential hematopoietic stem cell {MHSC}, from which all the cells of the circulating blood
are eventually derived.
The intermediatestage cells are very much like the multipotential stem cells, even though they
have already become committed to a particular line of cells; these are called committed stem
cells.
The hematopoiesis is driven by 2 factors:
1. Growth inducers (factors; e.g., The growth inducers promote growth but not
interleukin-3). differentiation of the ce
Causes one type of committed stem cell to differentiate
2. Differentiation inducers. one or more steps toward a final adult blood cell.
When both kidneys are removed from a person (anephric person), or when the
kidneys are destroyed by renal disease, the person invariably becomes very
anemic.
This is because the 10% of the normal erythropoietin formed in other
tissues (mainly in the liver) is sufficient to cause only one third to half the
RBC formation needed by the body.
Hematocrit (packed cell volume) ≈ 23–25% rather than 40–45%
When an animal or person is placed in an atmosphere of low oxygen, erythropoietin
begins to be formed within minutes to hours, and it reaches maximum production within
24 hours.
Yet, almost no new RBCs appear in the circulating blood until about 5 days later.
The rapid production of cells continues as long as the person remains in a low
oxygen state or until enough RBCs have been produced to carry adequate
amounts of oxygen to the tissues despite the low level of oxygen; at this time, the
rate of erythropoietin production decreases to a level that will maintain the
required number of RBCs but not an excess.
In the absence of erythropoietin, few RBCs are formed by the bone marrow.
At the other extreme, when large quantities of erythropoietin are formed, and if
plenty of iron and other required nutrients are available, the rate of RBC production
can rise to perhaps 10 or more times normal.
As would be expected, their maturation and rate of production are affected greatly by a
person’s nutritional status.
Deficiency of amino acids, iron or vitamins caused:
1) Anemia.
2) Vit B12 and Folic acid are essential for synthesis of DNA because each, in a different
way, is required for formation of thymidine triphosphate, one of the essential building
blocks of DNA.
3) Lack of vitamin B12 or folic acid causes abnormal and diminished DNA and,
consequently, failure of nuclear maturation and cell division.
4) Furthermore, the erythroblastic cells of the bone marrow, in addition to failing to
proliferate rapidly, produce mainly larger than normal RBCs called macrocytes, which
have a flimsy ( )رقيقmembrane and are often irregular, large, and oval instead of the usual
biconcave disc.
These poorly formed cells (macrocytes,), after entering the circulating blood, are
capable of carrying oxygen normally, but their fragility causes them to have a
short life, half to one-third normal. Therefore, deficiency of vitamin B12 or folic
acid causes maturation failure in the process of erythropoiesis.
Vitamin B12
Is a water-soluble vitamin that is naturally present in
some foods, added to others, and available as a
dietary supplement and a prescription medication.
Because vitamin B12 contains the mineral cobalt,
compounds with vitamin B12 activity are collectively
called “Cobalamins”.
The main dietary source is liver, kidney, red meat,
eggs, shellfish and dairy products.
Vitamin B12 is relatively stable and little is lost during
cooking.
B) Intestinal malabsorption.
The most common cause of the deficiency, which could be due to:
2) Gastrointestinal disease:
Total gastrectomy: the anaemia is developed after depletion of the body stores, which is usually,
occur within 5 years. This is sever when accompanied with iron deficiency anaemia.
Partial gastrectomy: (stagnant or blind loop syndrome) Partial removal of the stomach, and
refashioning the junction with the gut. The sterile duodenal part will colonized with bacteria, which
will consume huge amount of the vitamin.
3) Drug-induced Malabsorption:
Anticonvulsant, phenytoin.
Antimicrobial, neomycin.
Antigout, colchicine.
Alcohol.
C) Increased Requirements.
The requirements are increased during pregnancy. The increase is not sufficient to cause deficiency
unless the pregnant was previously borderline body stores of the vitamin.
Typical body stores of folate in a normal, healthy adult are about 10 mg and are
located in liver.
Thus, if dietary folate intake or intestinal absorption ceased, the body stores
would become exhausted in about 3-4 months.
The ideal diet contains 700 𝝁𝒈 of folate of which about half is absorbed.
Folate is very labile ( )حساسto heat; cooking can destroy up to 90% of folate in it.
Body stores are only sufficient for 3 months when dietary intake stop.
2- Intestinal malabsorption.
Coelic disease: Villous atrophy, which decreases iron and folate absorption.
Tropical sprue: Similar to coelic disease.
Crohn disease: Generalized malabsorption in the intestine.
3- Increased requirement.
Pregnancy; the daily requirement for folate can rise to 500μg in the 3ed trimester of pregnancy.
More than 60% of pregnant women have subnormal folate concentration.
This recently, demonstrated to be associated with neural tube defects.
The anaemia of chronic haemolytic conditions such as sickle cell anaemia frequently is
exacerbated by folate deficiency. Sever haemolytic conditions increases the rate of haemopoiesis
by a factor of 10, which cannot be met by dietary sources.
A number of rare enzyme deficiencies have been reported which cause impairment of folate
metabolism. Most of these are associated with megaloblastic changes and mental retardation.
Degeneration of the dorsal and lateral columns of the spinal cord are typical findings in sever
megaloblastic anemia due to deficiency of vitamin B12. The mechanism is not known yet.
Folic acid deficiency in pregnancy is associated with the incidence of neural tube defects such as
spina bifida and is also believed to lead to mild dementia and impairment of intellectual function.
Hemoglobin (the oxygen-binding protein of red blood cells)
Hemoglobin (Hb) is a major hemoprotein of human body.
Hb Function:
2) Hb serves in transport and exchange of gases (O2 and CO2) between lungs and tissues.
Respiration mechanism:
Hemoglobin in the body is to combine with oxygen in the lungs and then to
release this oxygen readily in the peripheral tissue capillaries, where the
gaseous tension of oxygen is much lower than in the lungs.
3) Hemoglobin Plays Role as Buffer:
Amount of Hb
Adult males: 13.5–17.5 𝒈/𝒅𝑳
Adult females: 12.5–16.5 𝒈/𝒅𝑳
Infant: 14–19 𝒈/𝒅𝑳
Hb Biosynthesis:
6.25 g Hb/day
Produced during stages of erythropoiesis in bone marrow.
Synthesis of Hb begins in Proerythroblast
Hb structure:
Tetrameric (quaternary) structure.
Hb consists of 4 polypeptide subunits.
Globular protein contain 4 allosteric sites.
There are several slight variations in the different subunit hemoglobin chains,
depending on the amino acid composition of the polypeptide portion.
The different types of chains are designated as alpha (α) chains, beta (β) chains, (γ)
gamma chains, and (δ) delta chains.
The most common form of hemoglobin in adults, hemoglobin A, is a combination
of two alpha chains and two beta chains (MW 64,458).
Both alpha and beta subunits of hemoglobin consist primarily of α-helical
Secondary structure and each subunit has a non-polypeptide component,
called heme with an iron atom that binds with oxygen.
Each of these hemes can bind loosely with one molecule of oxygen, making a
total of four molecules of oxygen (or eight oxygen atoms) that can be transported
by each hemoglobin molecule.
The types of hemoglobin chains in the hemoglobin molecule determine the
binding affinity of the hemoglobin for oxygen.
Abnormalities of the chains can alter the physical characteristics of the
hemoglobin molecule as well, for example; sickle cell anemia.
Globin Subunits:
Adult Hemoglobin has 4 Polypeptide chains 2α and 2β (identical pair).
globin chains Alpha globin chains Beta Globin chains
Composition 141 amino acids. 146 amino acids
MW 15,126 Daltons 15,866 Daltons
Biosynthesis-Expression α Globin gene on β Globin gene on
16th Chromosome. 11th chromosome.
The concentration of hemoglobin does not rise above this value because this is
the metabolic limit of the cell’s hemoglobin-forming mechanism.
Furthermore, in normal people, the percentage of hemoglobin is almost always
near the maximum in each cell. However, when hemoglobin formation is deficient,
the percentage of hemoglobin in the cells may fall considerably below this value,
and the volume of the RBC may also decrease because of diminished hemoglobin
to fill the cell.
In the case of lack of oxygen, new red blood cells are produced, not
hemoglobin.
What Is Heme? Metalloporphyrins are inhibitors of the rate-
limiting enzyme, heme oxygenase, in the
Prosthetic group of Hemoproteins. pathway of heme degradation leading to
Red color pigment. bilirubin production.
Metalloporphyrin.
Chemically heme is a Ferroprotoporphyrin Ferroprotoporphyrin is an iron-containing
cofactor.
Heme is a derivative of the porphyrin.
Porphyrins are cyclic compounds formed by fusion of 4 pyrrole rings
linked by methenyl bridges.
Most common porphyrin in humans is heme.
Iron in Heme:
Iron content of Hb: 3.4 mg / g of Hb
Fe of Heme is Hexavalent:
Fe of heme forms 6 coordinated bonds to satisfy its six
valencies:
4 bonds linked with each nitrogen of 4 pyrrole rings.
5th bond linked with Proximal Histidine.
6th bond is with Oxygen.
Iron-protoporphyrin-IX
Heme synthesishemoglobin:
The major sites of heme biosynthesis are the liver, which synthesizes a
number of heme proteins (particularly the CYP proteins}, and the erythrocyte-
producing cells of the bone marrow, which are active in Hb synthesis.
Over 85% of all heme synthesis occurs in erythroid tissue.
Mature red blood cells (RBC) lack mitochondria and are unable to
synthesize heme.
In the liver The rate of heme synthesis is highly variable caused by fluctuating
demands for hemeproteins.
in erythroid cells Relatively constant and is matched to the rate of globin synthesis.
Synthesis of heme:
Liver.
Bone Morrow.
No heme synthesis in RBC; don’t have mitochondria
Substrates mainly include succinyl-CoA, glycine, Fe2+.
Almost all the tissues can synthesize heme.
Major sites of synthes is liver and bone marrow
bone - marrow heme production equal to globin synthesis.
liver Variable dependent on heme pool balance.
ALA Synthase is the committed step (2 substeps) of the heme synthesis pathway,
& is usually rate limiting for the overall pathway.
Regulation occurs through control of gene transcription.
Mechanism
A) In the mitochondria Combination of Succinyl-CoA + Glycine in the process of
forming 𝜶-Amino-𝜷 −ketoadipate (Schiff base).
B) By decarboxylation using ALA synthase to produce ALA.
2. Hematin will also inhibit the translocation of ALA synthase from the cytoplasm
into the mitochondria where its substrate, succinyl CoA is formed. thus heme
synthesis is inhibited till there are sufficient globin chains to bind with.
Porphyria:
Porphyria is a name given to a group of metabolic disorders.
These disorders cause the individual to accumulate "porphyrins" or "porphyrin
precursors" in their body.
Which in turn causes an abundance of the porphyrins.
In porphyria, the cells do not convert porphyrins to heme in a normal manner,
which will result in the accumulation of the heme synthesis intermediate
products, related to as Porphyria.
The types that affect the nervous system are also known as acute porphyria, as
symptoms are rapid in onset and short in duration.
Symptoms of an attack include abdominal pain, chest pain, vomiting,
confusion, constipation, fever, high blood pressure, and high heart rate.
Porphyrias are genetic diseases in which activity of one of the enzymes involved
in heme synthesis is decreased (e.g., PBG Synthase, Porphobilinogen
Deaminase, etc…).
They are dominate, recessive autosomal and X-linked genes.
Symptoms of porphyria vary depending on:
The enzyme.
The severity of the deficiency (partial or complete).
Whether heme synthesis is affected primarily in liver or in developing
erythrocytes.
2) Photosensitivity:
Is another common symptom of porphyria.
Formation of superoxide radicals.
Skin damage may result from exposure to light.
This is attributable to elevated levels of
light-absorbing pathway intermediates and
their degradation products.
Symptoms include:
Skin rashes and blisters early in childhood.
cholestatic liver cirrhosis and progressive liver failure.
Lead poisoning
Inhibition of ferrochelatase, ALA dehydratase.
Displaces Zn+2 at enzyme active site.
Children:
Developmental defects.
Drop in IQ.
Hyperactivity.
Insomnia.
Many other health problems.
Adults:
Severe abdominal pain.
Mental confusion.
Many other symptoms.
Prehepatic (hemolytic) jaundice:
Results from excess production of bilirubin (beyond the livers ability to
conjugate it) following hemolysis.
Excess RBC lysis is commonly the result of autoimmune disease; hemolytic
disease of the newborn (Rh- or ABO incompatibility); structurally abnormal RBCs
(Sickle cell disease).
High plasma concentrations of unconjugated bilirubin (normal concentration
~0.5 mg/dL).
Intrahepatic jaundice
Impaired uptake, conjugation, or secretion of bilirubin.
Reflects a generalized liver (hepatocyte) dysfunction.
In this case, hyperbilirubinemia is usually accompanied by other abnormalities
in biochemical markers of liver function.
Posthepatic jaundice:
Caused by an obstruction of the biliary tree.
Plasma bilirubin is conjugated, and other biliary metabolites, such as bile acids
accumulate in the plasma.
Characterized by pale colored stools (absence of fecal bilirubin or urobilin),
and dark urine (increased conjugated bilirubin).
In a complete obstruction, urobilin is absent from the urine.
Neonatal Jaundice:
Common, particularly in premature infants.
Transient (resolves in the first 10 days).
Due to immaturity of the enzymes involved in bilirubin conjugation.
High levels of unconjugated bilirubin are toxic to the newborn – due to its
hydrophobicity it can cross the blood-brain barrier and cause a type of
mental retardation known as kernicterus.
If bilirubin levels are judged to be too high, then phototherapy with UV light
is used to convert it to a water soluble, non-toxic form.
IRON METABOLISM:
The body cannot get rid of iron except in two cases:
1) Shedding the epithelium.
2) The menstrual cycle; therefore, women are more susceptible to iron deficiency.
The total quantity of iron in the body averages 4 to 5 grams, about 65% of
which is in the form of hemoglobin.
4% myoglobin
1% heme compounds
0.1% combined with the protein transferrin in the blood
Stored for later mainly in the reticuloendothelial system
15% to 30% and liver parenchymal cells, principally in the form of
ferritin.
The iron is loosely bound in the transferrin and, consequently, can be released to
any tissue cell at any point in the body.
Excess iron in the blood is deposited especially in the liver hepatocytes and less
in the reticuloendothelial cells of the bone marrow.
In the cell cytoplasm, iron combines mainly with a protein, apoferritin, to form
ferritin.
Apoferritin has a molecular weight of about 460,000, and varying quantities of iron
can combine in clusters of iron radicals with this large molecule; therefore, ferritin
may contain only a small or a large amount of iron. This iron stored as ferritin is
called storage iron.
Smaller quantities of the iron in the storage pool are in an extremely insoluble
form called hemosiderin.
This is especially true when the total quantity of iron in the body is more
than the apoferritin storage pool can accommodate.
Hemosiderin large particles can be observed microscopically
ferritin So small particles Can be seen in the cell cytoplasm only
with an electron microscope.
Recycling of RBCs:
About 90% of the red blood cells are recycled by macrophages within the spleen,
liver and lymph nodes.
When RBCs have lived their life span of about 120 days and are destroyed,
the hemoglobin released from the cells is ingested by monocyte-
macrophage cells.
There, iron is liberated and is stored mainly in the ferritin pool to be used as
needed for the formation of new hemoglobin.
man woman
An average excretes about 0.6 mg of iron Additional menstrual loss of
each day, mainly into the feces. Additional blood brings long-term iron loss
quantities of iron are lost when bleeding to an average of about 1.3
occurs mg/day.
Absorption of Iron from the Intestinal Tract
Iron absorption from the intestines is extremely slow, at a maximum rate of
only a few milligrams per day. This slow rate of absorption means that even when
tremendous quantities of iron are present in the food, only small proportions can
be absorbed.
Iron is absorbed from all parts of the small intestine, mostly by the following
mechanism.
1) The liver secretes moderate amounts
of apotransferrin into the bile, which
flows through the bile duct into the
duodenum.
2) The apotransferrin binds with free iron
and also with certain iron compounds,
such as hemoglobin and myoglobin
(monomers) from meat, two of the most
important sources of iron in the diet.
This combination is called
transferrin.
3) In turn, it is attracted to and binds with
receptors in the membranes of intestinal
epithelial cells.
4) By pinocytosis, the transferrin
molecule, carrying its iron store, is
absorbed into the epithelial cells and
later released into the blood capillaries
beneath these cells in the form of plasma
transferrin.
Without hemoglobin; the heart would have to pump 140 liters per
minute Instead of normally 4 liters per minute.
Oxygenation/Loading of Oxygen
Hemoglobin gets oxygenated:
At lungs
At increased pO2 concentration (100-120 mm Hg) and decreased pCO2
Let us consider how the cooperative behavior indicated by the sigmoid curve
leads to efficient oxygen transport.
In the lungs, hemoglobin becomes nearly saturated with oxygen such that 98%
of the oxygen-binding sites are occupied.
When hemoglobin moves to the tissues and releases O2, the saturation
level drops to 32%.
Thus, a total of (98 – 32) = 66% of the potential oxygen-binding sites
contribute to oxygen transport.
If myoglobin were employed for oxygen transport, it would be 98% saturated in
the lungs, but would remain 91% saturated in the tissues.
Only (98 – 91) = 7% of the sites would contribute to oxygen transport.
The most oxygen that could be transported from a region in which pO2 is
100 torr to one in which it is 20 torr is (63 – 25) = 38%.
Embryonic Hb:
Hbε Gower-1 (ζ2ε2) Primary Hb in embryonic life ≈ 8 wks.
Portland-1 (ζ2γ2) is the ζ-substituted counterpart of fetal HbF (α2γ2) (By 12 weeks
of gestation, embryonic Hb changes into HbF.
Hb Portland-2 (ζ2β2) since it occurs only in cases of an extreme type of α-
thalassemia.
Hydrogen Ions and Carbon Dioxide Promote the Release of Oxygen: The Bohr Effect
The Bohr Effect describes hemoglobin's lower affinity for oxygen secondary to increases in the partial
pressure of carbon dioxide and/or decreased blood pH. This lower affinity, in turn, enhances the
unloading of oxygen into tissues to meet the oxygen demand of the tissue
According to local regulations (allosteric) there are two states:
1) R-state: 2) T-state:
Active Relaxed. Inactive Tense.
Haemoglobin VS leghaemoglobin
Haemoglobin Red colored protein present in Alpha = chromosome 16
red blood cells (erythrocytes). Beta = chromosome 11
leghaemoglobin Red pigment present in Alpha = chromosome 16
the root nodules of The others chains encoded by
leguminous plants and assists chromosome 11
in nitrogen fixation.
Hemoglobin electrophoresis
Is a blood test that can detect different types of hemoglobin.
The test can detect hemoglobin S, the form associated with sickle cell disease,
as well as other abnormal types of hemoglobin, such as hemoglobin C.
It can also be used to investigate thalassemias.
Procedure:
The test uses the principles of gel electrophoresis to separate out the various types of
hemoglobin and is a type of native gel electrophoresis.
After the sample has been treated to release the hemoglobin from the red cells, it is
introduced into a porous gel (usually made of agarose or cellulose acetate) and
subjected to an electrical field, most commonly in an alkaline medium.
Different hemoglobins have different charges, and according to those charges, they
move at different speeds in the gel and eventually form discrete bands.
A quality control sample containing hemoglobins A, F, S, and C is
run along with the patient sample to aid in identifying the different
bands.
The relative amounts of each type of hemoglobin can be estimated
by measuring the optical density of the bands, though this method is
not reliable for hemoglobins that are present in low quantities.
Clinical significance:
Adult human blood normally contains three types of hemoglobin:
Hemoglobin A, which makes up approximately 95% of the total.
Hemoglobin A2, which accounts for less than 3.5%; and a minute amount of
hemoglobin F.
If abnormal hemoglobin variants such as hemoglobin S, C or E are present, they will
appear as unexpected bands on electrophoresis (provided they do not migrate to the
same place as other hemoglobins).
Hemoglobin A2 levels are typically elevated in beta-thalassemia minor and
hemoglobin F may be slightly increased.
In beta-thalassemia major, hemoglobin A is decreased (or in some cases absent) and
hemoglobin F is markedly elevated; A2 levels are variable.
In hemoglobin H disease, a form of alpha-thalassemia, an abnormal band of
hemoglobin H can be detected, and sometimes a band of Hemoglobin Barts; but in the
milder alpha-thalassemia trait, electrophoresis results are effectively normal
2) White blood cells (leucocytes)
Classification of WBCs:
1) Granular leukocytes (polymorphnucleated):
2) agranular leukocytes:
3. LM appearance in smear:
4. Function:
Primarily antibacterial.
Neutrophils leave the blood and follow
chemotaxis signals to sites of wounding or other
inflammation, and phagocytose foreign agents
such as bacteria.
Pus is composed largely of dead neutrophils.
Eosinophil:
1. Life Span: < 2 weeks.
2. LM appearance in smear:
3. Function:
Anti-parasitic activity.
Mediators of inflammatory/allergic
responses in tissues.
Basophil
Lifespan: 1-2 years.
3. LM appearance in smear:
4. Function:
2. LM appearance in smear:
3. Function:
2. LM appearance in smears:
3. Function:
UNIT VI
Granulocytes, the Monocyte-Macrophage System,
and Inflammation
Our bodies are exposed continually to bacteria, viruses, eosinophils (polymorphonuclear), basophils (polymorpho-
fungi, and parasites, all of which occur normally and to nuclear), monocytes, lymphocytes and, occasionally, plasma
varying degrees in the skin, mouth, respiratory passage- cells. In addition, there are large numbers of platelets, which
ways, intestinal tract, lining membranes of the eyes, and are fragments of another type of cell similar to the WBCs
even the urinary tract. Many of these infectious agents found in the bone marrow, the megakaryocyte. The first three
are capable of causing serious abnormal physiological types of cells, the polymorphonuclear cells, all have a granu-
function or even death if they invade deeper tissues. We lar appearance, as shown in cell numbers 7, 10, and 12 in
are also exposed intermittently to other highly infectious Figure 34-1, and for this reason they are called granulocytes.
bacteria and viruses besides those that are normally pres- The granulocytes and monocytes protect the body
ent, and these agents can cause acute lethal diseases such against invading organisms by ingesting them (by phago-
as pneumonia, streptococcal infection, and typhoid fever. cytosis) or by releasing antimicrobial or inflammatory
Our bodies have a special system for combating the dif- substances that have multiple effects that aid in destroy-
ferent infectious and toxic agents. This system is composed ing the offending organism. The lymphocytes and plasma
of blood leukocytes (white blood cells [WBCs]) and tissue cells function mainly in connection with the immune
cells derived from leukocytes. These cells work together system, as discussed in Chapter 35. Finally, the function
in two ways to prevent disease: (1) by actually destroying of platelets is specifically to activate the blood-clotting
invading bacteria or viruses by phagocytosis; and (2) by mechanism, discussed in Chapter 37.
forming antibodies and sensitized lymphocytes that may
destroy or inactivate the invader. This chapter discusses the
first of these methods, and Chapter 35 discusses the second. Concentrations of Different White Blood Cells in
Blood. An adult human has about 7000 WBCs per mi-
croliter of blood (in comparison with 5 million red blood
LEUKOCYTES (WHITE BLOOD CELLS)
cells [RBCs] per microliter). Of the total WBCs, the nor-
The leukocytes, also called white blood cells, are the mobile mal percentages of the different types are approximately
units of the body’s protective system. They are formed the following:
partially in the bone marrow (granulocytes and monocytes • Neutrophils: 62.0%
and a few lymphocytes) and partially in the lymph tissue • Eosinophils: 2.3%
(lymphocytes and plasma cells). After formation, they are • Basophils: 0.4%
transported in the blood to different parts of the body • Monocytes: 5.3%
where they are needed. • Lymphocytes: 30.0%
The real value of WBCs is that most of them are specif- The number of platelets, which are only cell fragments,
ically transported to areas of serious infection and inflam- in each microliter of blood is normally between 150,000
mation, thereby providing a rapid and potent defense and 450,000, averaging about 300,000.!
against infectious agents. As we see later, the granulo-
cytes and monocytes have a special ability to “seek out
GENESIS OF WHITE BLOOD CELLS
and destroy” a foreign invader.
Early differentiation of the multipotential hematopoi-
etic stem cell into the different types of committed stem
GENERAL CHARACTERISTICS OF
cells was shown in Figure 33-2 in the previous chapter.
LEUKOCYTES
Aside from the cells committed to form RBCs, two major
Types of White Blood Cells. Six types of WBCs are nor- lineages of WBCs are formed, the myelocytic and lym-
mally present in the blood: neutrophils (polymorphonuclear), phocytic lineages. The left side of Figure 34-1 shows the
449
UNIT VI Blood Cells, Immunity, and Blood Coagulation
3
2
13
4 8 11
14
5
Figure 34-1. Genesis of white blood
cells. The different cells of the myelo- 9
cyte series are shown: 1, myeloblast;
15
2, promyelocyte; 3, megakaryocyte; 4,
6
neutrophil myelocyte; 5, young neutro-
phil metamyelocyte; 6, band neutrophil
metamyelocyte; 7, neutrophil; 8, eo-
sinophil myelocyte; 9, eosinophil meta- 10 12 16
myelocyte; 10, eosinophil; 11, basophil 7
myelocyte; 12, basophil; 13–16, stages
of monocyte formation.
myelocytic lineage, beginning with the myeloblast; the needed. In times of serious tissue infection, this total life
right side shows the lymphocytic lineage, beginning with span is often shortened to only a few hours because the
the lymphoblast. granulocytes proceed even more rapidly to the infected
The granulocytes and monocytes are formed only area, perform their functions, and in the process, are
in the bone marrow. Lymphocytes and plasma cells are themselves destroyed.
produced mainly in the various lymphogenous tissues— The monocytes also have a short transit time, 10 to 20
especially the lymph glands, spleen, thymus, tonsils, and hours in the blood, before wandering through the cap-
various pockets of lymphoid tissue elsewhere in the body, illary membranes into the tissues. Once in the tissues,
such as in the bone marrow and in Peyer’s patches under- they swell to much larger sizes to become tissue macro-
neath the epithelium in the gut wall. phages and, in this form, they can live for months unless
The WBCs formed in the bone marrow are stored in destroyed while performing phagocytic functions. These
the marrow until they are needed in the circulatory sys- tissue macrophages are the basis of the tissue macrophage
tem. Then, when the need arises, various factors cause system (discussed in greater detail later), which provides
them to be released (these factors are discussed later). continuing defense against infection.
Normally, about three times as many WBCs are stored in Lymphocytes enter the circulatory system continually,
the marrow as circulate in the entire blood. This quantity along with drainage of lymph from the lymph nodes and
represents about a 6-day supply of these cells. other lymphoid tissue. After a few hours, they pass out
The lymphocytes are mostly stored in the various lym- of the blood back into the tissues by diapedesis/extrava-
phoid tissues, except for a small number that are tempo- sation. Then, they re-enter the lymph and return to the
rarily being transported in the blood. blood again and again; thus, there is continual circulation
As shown in Figure 34-1, megakaryocytes (cell 3) are of lymphocytes through the body. Lymphocytes have life
also formed in the bone marrow. These megakaryocytes spans of weeks or months, depending on the body’s need
fragment in the bone marrow and the small fragments, for these cells.
known as platelets (or thrombocytes), then pass into the The platelets in the blood are replaced about once
blood. They are very important in the initiation of blood every 10 days. In other words, about 30,000 platelets are
clotting.! formed each day for each microliter of blood.!
450
Chapter 34 Resistance of the Body to Infection
UNIT VI
permeability
eral reaction products caused by plasma clotting in the
inflamed area, as well as other substances.
As shown in Figure 34-2, chemotaxis depends on the
concentration gradient of the chemotactic substance. The
concentration is greatest near the source, which directs
the unidirectional movement of the WBCs. Chemotaxis
Chemotaxis is effective up to 100 micrometers away from an inflamed
source
tissue. Therefore, because almost no tissue area is more
than 50 micrometers away from a capillary, the chemo-
tactic signal can easily move hordes of WBCs from the
capillaries into the inflamed area.!
451
UNIT VI Blood Cells, Immunity, and Blood Coagulation
452
Chapter 34 Resistance of the Body to Infection
Primary
nodule Capsule
Subcapsular
UNIT VI
sinus
Valve
Lymph in
medullary
sinuses
Germinal
center
Hilus Medullary cord
Efferent lymphatics
Figure 34-4. Functional diagram of a lymph node.
453
UNIT VI Blood Cells, Immunity, and Blood Coagulation
454
Chapter 34 Resistance of the Body to Infection
Receptors
Neutrophil
UNIT VI
Endothelial cell
Selectin ICAM-1
Cytokines
Inflamed tissue
Figure 34-7. Migration of neutrophils from the blood into inflamed tissue. Cytokines and other biochemical products of the inflamed tissue
cause increased expression of selectins and intercellular adhesion molecule-1 (ICAM-1) on the surface of endothelial cells. These adhesion mol-
ecules bind to complementary molecules or receptors on the neutrophil, causing it to adhere to the wall of the capillary or venule. The neutrophil
then migrates through the vessel wall by diapedesis or extravasation toward the site of tissue injury.
1. They cause increased expression of adhesion mol- neutrophils in the blood sometimes increases fourfold
ecules, such as selectins and intercellular adhesion to fivefold—from a normal of 4,000 to 5,000 to 15,000 to
molecule-1 (ICAM-1) on the surface of endothelial 25,000 neutrophils/µl. This is called neutrophilia, which
cells in the capillaries and venules. These adhesion means an increase in the number of neutrophils in the
molecules, reacting with complementary integrin blood. Neutrophilia is caused by products of inflamma-
molecules on the neutrophils, cause the neutro- tion that enter the blood stream, are transported to the
phils to stick to the capillary and venule walls in bone marrow, and act there on the stored neutrophils of
the inflamed area. This effect is called margination the marrow to mobilize these into the circulating blood.
and is shown in Figure 34-2 and in more detail in This makes even more neutrophils available to the in-
Figure 34-7. flamed tissue area.!
2. They also cause the intercellular attachments be-
tween the endothelial cells of the capillaries and Second Macrophage Invasion Into the Inflamed
small venules to loosen, allowing openings large Tissue Is a Third Line of Defense. Along with the in-
enough for neutrophils to crawl through the capil- vasion of neutrophils, monocytes from the blood enter
laries by diapedesis into the tissue spaces. the inflamed tissue and enlarge to become macrophages.
3. They then cause chemotaxis of the neutrophils to- However, the number of monocytes in the circulating
ward the injured tissues, as explained earlier. The blood is low. Also, the storage pool of monocytes in the
entire process of neutrophil (or other substances bone marrow is much less than that of neutrophils. There-
and cells such as monocytes) translocation through fore, the buildup of macrophages in the inflamed tissue
the capillaries into the tissues surrounding them is area is much slower than that of neutrophils, requiring
called extravasation; the specific passage of blood several days to become effective. Furthermore, even after
cells through the intact walls of the capillaries is invading the inflamed tissue, monocytes are still immature
called diapedesis, although this term is often used cells, requiring 8 hours or more to swell to much larger siz-
interchangeably with extravasation when discussing es and develop tremendous quantities of lysosomes. Only
blood cell movement through the capillaries into then do they acquire the full capacity of tissue macrophages
tissues. for phagocytosis. After several days to several weeks, the
Thus, within several hours after tissue damage begins, macrophages finally come to dominate the phagocytic
the area becomes well supplied with neutrophils. Because cells of the inflamed area because of greatly increased bone
the blood neutrophils are already mature cells, they are marrow production of new monocytes, as explained later.
ready to begin their scavenger functions of killing bacteria As already noted, macrophages can phagocytize far
and removing foreign matter immediately.! more bacteria (about five times as many) and far larger
particles, including even neutrophils and large quantities
Acute Increase in the Number of Neutrophils in of necrotic tissue, than can neutrophils. Also, the macro-
Blood—Neutrophilia. Also, within a few hours after phages play an important role in initiating development of
the onset of acute severe inflammation, the number of antibodies, as discussed in Chapter 35.!
455
UNIT VI Blood Cells, Immunity, and Blood Coagulation
456
Chapter 34 Resistance of the Body to Infection
Eosinophils also have a special propensity to collect in always find bacteria on the surfaces of the eyes, urethra,
tissues in which allergic reactions occur, such as in the and vagina. Any decrease in the number of WBCs imme-
peribronchial tissues of the lungs in people with asthma diately allows invasion of adjacent tissues by bacteria that
and in the skin after an allergic skin reaction. This action are already present.
is caused at least partly by the fact that many mast cells Within 2 days after the bone marrow stops producing
UNIT VI
and basophils participate in allergic reactions, as dis- WBCs, ulcers may appear in the mouth and colon, or some
cussed in the next paragraph. The mast cells and baso- form of severe respiratory infection might develop. Bacte-
phils release an eosinophil chemotactic factor that causes ria from the ulcers rapidly invade surrounding tissues and
eosinophils to migrate toward the inflamed allergic tis- the blood. Without treatment, death often ensues in less
sue. The eosinophils are believed to detoxify some of the than 1 week after acute total leukopenia begins.
inflammation-inducing substances released by the mast Irradiation of the body by x-rays or gamma rays, or
cells and basophils and probably also phagocytize and exposure to drugs and chemicals that contain benzene or
destroy allergen-antibody complexes, thus preventing anthracene nuclei, is likely to cause aplasia of the bone
excess spread of the local inflammatory process.! marrow. Some common drugs such as chloramphenicol
(an antibiotic), thiouracil (used to treat thyrotoxicosis),
and even various barbiturate hypnotics on rare occasions
BASOPHILS
cause leukopenia, thus setting off the entire infectious
The basophils in the circulating blood are similar to the sequence of this disorder.
large tissue mast cells located immediately outside many After moderate irradiation injury to the bone marrow,
of the capillaries in the body. Both mast cells and baso- some stem cells, myeloblasts, and hemocytoblasts may
phils liberate heparin into the blood. Heparin is a sub- remain undestroyed in the marrow and are capable of
stance that can prevent blood coagulation. regenerating the bone marrow, provided sufficient time
The mast cells and basophils also release histamine, as is available. A patient properly treated with transfusions,
well as smaller quantities of bradykinin and serotonin. It is plus antibiotics and other drugs to ward off infection, usu-
mainly the mast cells in inflamed tissues that release these ally develops enough new bone marrow within weeks to
substances during inflammation. months for blood cell concentrations to return to normal.!
The mast cells and basophils play an important role
in some types of allergic reactions because the type of
LEUKEMIAS
antibody that causes allergic reactions, immunoglobulin
E (IgE), has a special propensity to become attached to Uncontrolled production of WBCs can be caused by can-
mast cells and basophils. Then, when the specific anti- cerous mutation of a myelogenous or lymphogenous cell.
gen for the specific IgE antibody subsequently reacts This process causes leukemia, which is usually character-
with the antibody, the resulting attachment of antigen ized by greatly increased numbers of abnormal WBCs in
to antibody causes the mast cell or basophil to release the circulating blood.
increased quantities of histamine, bradykinin, sero- There are two general types of leukemia, lymphocytic
tonin, heparin, slow-reacting substance of anaphylaxis and myelogenous. The lymphocytic leukemias are caused
(a mixture of three leukotrienes), and several lysosomal by cancerous production of lymphoid cells, usually begin-
enzymes. These substances cause local vascular and ning in a lymph node or other lymphocytic tissue and
tissue reactions that mediate many, if not most, of the spreading to other areas of the body. The second type of
allergic manifestations. These reactions are discussed in leukemia, myelogenous leukemia, begins by cancerous
greater detail in Chapter 35.! production of young myelogenous cells in the bone mar-
row and then spreads throughout the body so that WBCs
are produced in many extramedullary tissues—especially
LEUKOPENIA
in the lymph nodes, spleen, and liver.
A clinical condition known as leukopenia, in which In myelogenous leukemia, the cancerous process occa-
the bone marrow produces very few WBCs, occasion- sionally produces partially differentiated cells, resulting in
ally occurs. This condition leaves the body unprotected what might be called neutrophilic leukemia, eosinophilic
against many bacteria and other agents that might invade leukemia, basophilic leukemia, or monocytic leukemia.
the tissues. More frequently, however, the leukemia cells are bizarre
Normally, the human body lives in symbiosis with and undifferentiated and not identical to any of the nor-
many bacteria because the mucous membranes of the mal WBCs. Usually, the more undifferentiated the cell, the
body are constantly exposed to large numbers of bac- more acute is the leukemia, often leading to death within a
teria. The mouth almost always contains various spiro- few months if untreated. With some of the more differen-
chetal, pneumococcal, and streptococcal bacteria, and tiated cells, the process can be chronic, sometimes devel-
these same bacteria are present to a lesser extent in the oping slowly over 10 to 20 years. Leukemic cells, especially
entire respiratory tract. The distal gastrointestinal tract is the very undifferentiated cells, are usually nonfunctional
especially loaded with colon bacilli. Furthermore, one can for providing normal protection against infection.
457
UNIT VI Blood Cells, Immunity, and Blood Coagulation
Effects of Leukemia on the Body Hallek M, Shanafelt TD, Eichhorst B: Chronic lymphocytic leukaemia.
Lancet 391:1524, 2018.
The first effect of leukemia is metastatic growth of leuke- Honda M, Kubes P: Neutrophils and neutrophil extracellular traps in
mic cells in abnormal areas of the body. Leukemic cells the liver and gastrointestinal system. Nat Rev Gastroenterol Hepa-
from the bone marrow may reproduce so much that they tol 15:206, 2018.
Lemke G: How macrophages deal with death. Nat Rev Immunol 19:
invade the surrounding bone, causing pain and, eventu-
539, 2019.
ally, a tendency for bones to fracture easily. Liew PX, Kubes P: The neutrophil’s role during health and disease.
Almost all leukemias eventually spread to the spleen, Physiol Rev 99:1223, 2019.
lymph nodes, liver, and other vascular regions, regardless Medzhitov R: Origin and physiological roles of inflammation. Nature
of whether the leukemia originated in the bone marrow or 454:428, 2008.
Ng LG, Ostuni R, Hidalgo A: Heterogeneity of neutrophils. Nat Rev
lymph nodes. Common effects in leukemia are the develop-
Immunol 19:255, 2019.
ment of infection, severe anemia, and a bleeding tendency Papayannopoulos V: Neutrophil extracellular traps in immunity and
caused by thrombocytopenia (lack of platelets). These effects disease. Nat Rev Immunol 18:134, 2018.
result mainly from displacement of the normal bone mar- Phillipson M, Kubes P: The healing power of neutrophils. Trends
row and lymphoid cells by the nonfunctional leukemic cells. Immunol 2019 May 31. pii: S1471-4906(19)30103-30106.
Pinho S, Frenette PS: Haematopoietic stem cell activity and interac-
Finally, an important effect of leukemia on the body
tions with the niche. Nat Rev Mol Cell Biol 20:303, 2019.
is excessive use of metabolic substrates by the growing Russell DG, Huang L, VanderVen BC: Immunometabolism at the
cancerous cells. The leukemic tissues reproduce new interface between macrophages and pathogens. Nat Rev Immunol
cells so rapidly that tremendous demands are made on 19:291, 2019.
the body reserves for foodstuffs, specific amino acids, Short NJ, Rytting ME, Cortes JE: Acute myeloid leukaemia. Lancet
392:593, 2018.
and vitamins. Consequently, the energy of the patient is
Spivak JL: Myeloproliferative neoplasms. N Engl J Med 376:2168,
greatly depleted, and excessive utilization of amino acids 2017.
by leukemic cells causes especially rapid deterioration of Watanabe S, Alexander M, Misharin AV, Budinger GRS: The role of
the normal protein tissues of the body. Thus, while the macrophages in the resolution of inflammation. J Clin Invest 129:
leukemic tissues grow, other tissues become debilitated. 2619, 2019.
Werner S, Grose R: Regulation of wound healing by growth factors
After metabolic starvation has continued long enough,
and cytokines. Physiol Rev 83:835, 2003.
this factor alone is sufficient to cause death.
Bibliography
David BA, Kubes P: Exploring the complex role of chemokines and
chemoattractants in vivo on leukocyte dynamics. Immunol Rev
289:9, 2019.
DeNardo DG, Ruffell B: Macrophages as regulators of tumour immu-
nity and immunotherapy. Nat Rev Immunol 19:369, 2019.
458
CHAPTER 37
UNIT VI
Hemostasis and Blood Coagulation
HEMOSTASIS EVENTS fragment into the minute platelets in the bone marrow or
soon after entering the blood, especially as they squeeze
The term hemostasis means prevention of blood loss. through capillaries. The normal concentration of platelets
Whenever a vessel is severed or ruptured, hemostasis is in the blood is between 150,000 and 450,000/µl.
achieved by several mechanisms: (1) vascular constric- Platelets have many functional characteristics of whole
tion; (2) formation of a platelet plug; (3) formation of a cells, even though they do not have nuclei and cannot
blood clot as a result of blood coagulation; and (4) even- reproduce. In their cytoplasm are the following: (1) actin
tual growth of fibrous tissue into the blood clot to close and myosin molecules, which are contractile proteins
the hole in the vessel permanently. similar to those found in muscle cells, and still another
VASCULAR CONSTRICTION contractile protein, thrombosthenin, that can cause
the platelets to contract; (2) residuals of both the endo-
Immediately after a blood vessel has been cut or rup- plasmic reticulum and Golgi apparatus that synthesize
tured, the trauma to the vessel wall causes smooth muscle various enzymes and especially store large quantities of
in the wall to contract; this instantaneously reduces the calcium ions; (3) mitochondria and enzyme systems that
flow of blood from the ruptured vessel. The contraction are capable of forming adenosine triphosphate (ATP) and
results from the following: (1) local myogenic spasm; (2) adenosine diphosphate (ADP); (4) enzyme systems that
local autacoid factors from the traumatized tissues, vas- synthesize prostaglandins, which are local hormones that
cular endothelium, and blood platelets; and (3) nervous cause many vascular and other local tissue reactions; (5)
reflexes. The nervous reflexes are initiated by pain nerve an important protein called fibrin-stabilizing factor, which
impulses or other sensory impulses that originate from the we discuss later in relation to blood coagulation; and (6)
traumatized vessel or nearby tissues. However, even more a growth factor that causes vascular endothelial cells, vas-
vasoconstriction probably results from local myogenic cular smooth muscle cells, and fibroblasts to multiply and
contraction of the blood vessels initiated by direct dam- grow, thus causing cellular growth that eventually helps
age to the vascular wall. And, for the smaller vessels, the repair damaged vascular walls.
platelets are responsible for much of the vasoconstriction On the platelet cell membrane surface is a coat of glyco-
by releasing a vasoconstrictor substance, thromboxane A2. proteins that repulses adherence to normal endothelium
The more severely a vessel is traumatized, the greater and yet causes adherence to injured areas of the vessel
the degree of vascular spasm. The spasm can last for many wall, especially to injured endothelial cells and even more
minutes or even hours, during which time the processes so to any exposed collagen from deep within the vessel
of platelet plugging and blood coagulation can take place.! wall. In addition, the platelet membrane contains large
amounts of phospholipids that activate multiple stages in
FORMATION OF THE PLATELET PLUG
the blood-clotting process, as discussed later.
If the cut in the blood vessel is very small—many very Thus, the platelet is an active structure. It has a half-life
small vascular holes develop throughout the body each in the blood of only 8 to 12 days, so over several weeks its
day—the cut is often sealed by a platelet plug rather than functional processes run out; it is then eliminated from
by a blood clot. To understand this process, it is important the circulation mainly by the tissue macrophage system.
that we first discuss the nature of platelets themselves. More than half of the platelets are removed by macro-
phages in the spleen, where the blood passes through a
Physical and Chemical Characteristics latticework of tight trabeculae.!
Platelets (also called thrombocytes) are minute discs 1 to
4 micrometers in diameter. They are formed in the bone Mechanism of Platelet Plug Formation
marrow from megakaryocytes, which are extremely large Platelet repair of vascular openings is based on several
hematopoietic cells in the marrow; the megakaryocytes important functions of the platelet. When platelets
477
UNIT VI Blood Cells, Immunity, and Blood Coagulation
when platelet
come in contact with a damaged vascular surface,
especially with collagen fibers in the vascular wall, the
platelets rapidly change their own characteristics dras-
tically (Figure 37-1). They begin to swell, they assume
irregular forms with numerous irradiating pseudo- 1. Severed vessel 2. Platelets agglutinate
pods protruding from their surfaces, their contractile
proteins contract forcefully and cause the release of
granules that contain multiple active factors, and they
become sticky so that they adhere to collagen in the
tissues and to a protein called von Willebrand factor 3. Fibrin appears 4. Fibrin clot forms
(vWF), which leaks into the traumatized tissue from
the plasma. The platelet surface glycoproteins bind to
vWF in the exposed matrix below the damaged endo-
thelium. The platelets then secrete increased quanti-
ties of ADP and platelet- activating factor (PAF), and
their enzymes form thromboxane A2. Thromboxane is 5. Clot retraction occurs
a vasoconstrictor and, along with ADP and PAF, acts on Figure 37-2. Clotting process in a traumatized blood vessel. (Modi-
nearby platelets to activate them as well; the stickiness fied from Seegers WH: Hemostatic Agents. Springfield, IL: Charles C
of these additional activated platelets causes them to Thomas, 1948.)
adhere to the original activated platelets.
Therefore, at the site of a puncture in a blood vessel
wall, the damaged vascular wall activates successively BLOOD COAGULATION IN THE RUPTURED
increasing numbers of platelets that attract more and VESSEL
more additional platelets, thus forming a platelet plug. The third mechanism for hemostasis is formation of the
This plug is loose at first but is usually successful in block- blood clot. The clot begins to develop in 15 to 20 seconds
ing blood loss if the vascular opening is small. Then, dur- if the trauma to the vascular wall is severe and in 1 to 2
ing the subsequent process of blood coagulation, fibrin minutes if the trauma is minor. Activator substances from
threads form. These threads attach tightly to the platelets, the traumatized vascular wall, from platelets, and from
thus constructing an unyielding plug. blood proteins adhering to the traumatized vascular wall
Importance of Platelet Mechanism for Closing initiate the clotting process. The physical events of this
Vascular Holes. The platelet- plugging mechanism is process are shown in Figure 37-2; Table 37-1 lists the
extremely important for closing minute ruptures in most important clotting factors.
very small blood vessels that occur many thousands of Within 3 to 6 minutes after rupture of a vessel, the
times daily. Indeed, multiple small holes through the entire opening or broken end of the vessel is filled with
endothelial cells themselves are often closed by plate- clot if the vessel opening is not too large. After 20 to 60
lets actually fusing with the endothelial cells to form minutes, the clot retracts, which closes the vessel still
additional endothelial cell membranes. Literally thou- further. Platelets also play an important role in this clot
sands of small hemorrhagic areas develop each day un- retraction, as discussed later.!
der the skin (petechiae, which appear as purple or red
FIBROUS ORGANIZATION OR
dots on the skin) and throughout the internal tissues
DISSOLUTION OF BLOOD CLOTS
of a person who has few blood platelets. This phenom-
enon does not occur in persons with normal numbers Once a blood clot has formed, it can follow one of two
of platelets.! courses: (1) it can become invaded by fibroblasts, which
478
Chapter 37 Hemostasis and Blood Coagulation
UNIT VI
Tissue factor Factor III; tissue thromboplastin
Fibrinogen Fibrin monomer
Calcium Factor IV
Ca2+
Factor V Proaccelerin; labile factor; Ac-
globulin (Ac-G) Fibrin fibers
479
UNIT VI Blood Cells, Immunity, and Blood Coagulation
Prothrombin Thrombin
Cross-linked
Figure 37-4. Coagulation cascade after Fibrinogen Fibrin
Platelet fibrin
vascular injury. Exposure of blood to the
Release of phospholipid
vascular wall causes release of tissue factor
tissue factor complex
(also called factor III or thromboplastin) from
endothelial cells, phospholipid expression,
activation of thrombin, which then acts on
fibrinogen to form fibrin, and fibrin polym-
erization to form a meshwork that stabilizes
the platelet plug. Endothelium Fibrin clot
Platelets also play an important role in the conversion Action of Thrombin on Fibrinogen to Form Fibrin.
of prothrombin to thrombin because much of the pro- Thrombin is a protein enzyme with weak proteolytic
thrombin first attaches to prothrombin receptors on the capabilities. It acts on fibrinogen to remove four low-
platelets that are already bound to the damaged tissue. molecular-weight peptides from each molecule of fi-
brinogen, forming one molecule of fibrin monomer that
Prothrombin and Thrombin. Prothrombin is a plasma
has the automatic capability to polymerize with other fi-
protein, an α2-globulin, having a molecular weight of
brin monomer molecules to form fibrin fibers. Therefore,
68,700. It is present in normal plasma in a concentration
many fibrin monomer molecules polymerize within sec-
of about 15 mg/dl. It is an unstable protein that can split
onds into long fibrin fibers that constitute the reticulum of
easily into smaller compounds, one of which is thrombin,
the blood clot.
which has a molecular weight of 33,700, almost half that
In the early stages of polymerization, the fibrin mono-
of prothrombin.
mer molecules are held together by weak noncovalent
Prothrombin is formed continually by the liver, and it
hydrogen bonding, and the newly forming fibers are not
is continually being used throughout the body for blood
cross-linked with one another. Therefore, the resultant
clotting. If the liver fails to produce prothrombin, in a day
clot is weak and can be broken apart with ease. However,
or so prothrombin concentration in the plasma falls too
another process occurs during the next few minutes that
low to provide normal blood coagulation.
greatly strengthens the fibrin reticulum. This process
Vitamin K is required by the liver for normal activa-
involves a substance called fibrin stabilizing factor that
tion of prothrombin, as well as a few other clotting fac-
is present in small amounts in normal plasma globulins
tors. Therefore, lack of vitamin K or the presence of liver
but is also released from platelets entrapped in the clot.
disease that prevents normal prothrombin formation can
Before fibrin stabilizing factor can have an effect on the
decrease the prothrombin to such a low level that a bleed-
fibrin fibers, it must be activated. The same thrombin that
ing tendency results.!
causes fibrin formation also activates the fibrin stabiliz-
ing factor. This activated substance then operates as an
enzyme to form covalent bonds between more and more
CONVERSION OF FIBRINOGEN TO
of the fibrin monomer molecules, as well as multiple
FIBRIN—FORMATION OF THE CLOT
cross-linkages between adjacent fibrin fibers, thus adding
Fibrinogen Formed in the Liver Essential for Clot For- tremendously to the three-dimensional strength of the
mation. Fibrinogen is a high-molecular-weight protein fibrin meshwork.!
(molecular weight ≈340,000) that occurs in the plasma in
Blood Clot. The clot is composed of a meshwork of fi-
quantities of 100 to 700 mg/dl. Fibrinogen is formed in
brin fibers running in all directions and entrapping blood
the liver, and liver disease can decrease the concentration
cells, platelets, and plasma (see Figure 37-4). The fibrin
of circulating fibrinogen, as it does the concentration of
fibers also adhere to damaged surfaces of blood vessels;
prothrombin, noted earlier.
therefore, the blood clot becomes adherent to any vascu-
Because of its large molecular size, little fibrinogen
lar opening and thereby prevents further blood loss.!
normally leaks from the blood vessels into the intersti-
tial fluids, and because fibrinogen is one of the essential Clot Retraction and Expression of Serum. Within a few
factors in the coagulation process, interstitial fluids ordi- minutes after a clot is formed, it begins to contract and
narily do not coagulate. Yet, when the permeability of the usually expresses most of the fluid from the clot within
capillaries becomes pathologically increased, fibrinogen 20 to 60 minutes. The fluid expressed is called serum be-
does leak into the tissue fluids in sufficient quantities to cause all its fibrinogen and most of the other clotting fac-
allow clotting of these fluids in much the same way that tors have been removed; in this way, serum differs from
plasma and whole blood can clot.! plasma and cannot clot because it lacks these factors.
480
Chapter 37 Hemostasis and Blood Coagulation
Platelets are necessary for clot retraction to occur. (1) Tissue trauma
Therefore, failure of clot retraction is an indication that the
number of platelets in the circulating blood might be low.
Electron micrographs of platelets in blood clots show that
they become attached to the fibrin fibers in such a way that Tissue factor
UNIT VI
they actually bond different fibers together. Furthermore,
platelets entrapped in the clot continue to release proco-
agulant substances, one of the most important of which (2) Vll VIIa
is fibrin stabilizing factor, which causes more and more
cross-linking bonds between adjacent fibrin fibers. In addi- X Activated X (Xa)
tion, the platelets contribute directly to clot contraction
by activating platelet thrombosthenin, actin, and myosin Ca2+
molecules, which are all contractile proteins in the plate- V Ca2+
lets; they cause strong contraction of the platelet spicules
(3) Prothrombin
attached to the fibrin. This action also helps compress the activator
Platelet
fibrin meshwork into a smaller mass. The contraction is phospholipids
activated and accelerated by thrombin and by calcium ions
Prothrombin Thrombin
released from calcium stores in the mitochondria, endo-
plasmic reticulum, and Golgi apparatus of the platelets.
As the clot retracts, the edges of the broken blood ves- Ca2+
sel are pulled together, thus contributing still further to Figure 37-5. Extrinsic pathway for initiating blood clotting.
hemostasis.!
481
UNIT VI Blood Cells, Immunity, and Blood Coagulation
and, in the presence of calcium ions, acts enzymati- Intrinsic Pathway for Initiating Clotting
cally on factor X to form activated factor X (Xa). The second mechanism for initiating formation of pro-
3. Effect of Xa to form prothrombin activator—role thrombin activator, and therefore for initiating clotting,
of factor V. The activated factor X combines im- begins with trauma to the blood or exposure of the blood
mediately with tissue phospholipids that are part to collagen from a traumatized blood vessel wall. Then the
of tissue factors or with additional phospholipids process continues through the series of cascading reac-
released from platelets, as well as with factor V, tions shown in Figure 37-6.
to form the complex called prothrombin activator. 1. Blood trauma causes (1) activation of factor XII and
Within a few seconds, in the presence of Ca2+, pro- (2) release of platelet phospholipids. Trauma to the
thrombin is split to form thrombin, and the clot- blood or exposure of the blood to vascular wall col-
ting process proceeds as already explained. At first, lagen alters two important clotting factors in the
the factor V in the prothrombin activator complex blood: factor XII and the platelets. When factor XII
is inactive, but once clotting begins and thrombin is disturbed, such as by coming into contact with
begins to form, the proteolytic action of thrombin collagen or with a wettable surface such as glass, it
activates factor V. This activation then becomes an takes on a new molecular configuration that con-
additional strong accelerator of prothrombin ac- verts it into a proteolytic enzyme called activated
tivation. Thus, in the final prothrombin activator factor XII. Simultaneously, the blood trauma also
complex, activated factor X is the actual protease damages the platelets because of adherence to col-
that causes splitting of prothrombin to form throm- lagen or to a wettable surface (or by damage in other
bin. Activated factor V greatly accelerates this pro- ways); this releases platelet phospholipids that con-
tease activity, and platelet phospholipids act as a tain the lipoprotein called platelet factor 3, which
vehicle that further accelerates the process. Note also plays a role in subsequent clotting reactions.
especially the positive feedback effect of thrombin, 2. Activation of factor XI. The activated factor XII also
acting through factor V, to accelerate the entire pro- acts enzymatically on factor XI to activate this fac-
cess once it begins.! tor, which is the second step in the intrinsic path-
Blood trauma or
contact with collagen
(5) Platelet
phospholipids
Thrombin Ca2+
Prothrombin
activator
Platelet
phospholipids
Prothrombin Thrombin
482
Chapter 37 Hemostasis and Blood Coagulation
way. This reaction also requires high-molecular- the blood. With severe tissue trauma, clotting can occur
weight kininogen and is accelerated by prekallikrein. in as little as 15 seconds. The intrinsic pathway is much
3. Activation of factor IX by activated factor XI. The slower to proceed, usually requiring 1 to 6 minutes to
activated factor XI then acts enzymatically on fac- cause clotting.!
tor IX to activate this factor as well.
Intravascular Anticoagulants Prevent Blood
UNIT VI
4. Activation of factor X—role of factor VIII. The acti-
vated factor IX, acting in concert with activated fac- Clotting in the Normal Vascular System
tor VIII and the platelet phospholipids and factor III Endothelial Surface Factors. Probably the most impor-
from the traumatized platelets, activates factor X. It tant factors for preventing clotting in the normal vas-
is clear that when either factor VIII or platelets are cular system are the following: (1) the smoothness of the
in short supply, this step is deficient. Factor VIII is endothelial cell surface, which prevents contact activation
the factor that is missing in a person who has clas- of the intrinsic clotting system; (2) a layer of glycocalyx on
sic hemophilia, so it is called antihemophilic factor. the endothelium (glycocalyx is a mucopolysaccharide ad-
Platelets are the clotting factor that is lacking in the sorbed to the surfaces of the endothelial cells), which repels
bleeding disease called thrombocytopenia. clotting factors and platelets, thereby preventing activation
5. Action of activated factor X to form prothrombin of clotting; and (3) a protein bound with the endothelial
activator—role of factor V. This step in the intrinsic membrane, thrombomodulin, which binds thrombin. Not
pathway is the same as the last step in the extrinsic only does the binding of thrombin with thrombomodulin
pathway. That is, activated factor X combines with slow the clotting process by removing thrombin, but the
factor V and platelet or tissue phospholipids to form thrombomodulin-thrombin complex also activates a plas-
the complex called prothrombin activator. The pro- ma protein, protein C, that acts as an anticoagulant by inac-
thrombin activator, in turn, initiates the cleavage tivating activated factors V and VIII.
of prothrombin to form thrombin within seconds, When the endothelial wall is damaged, its smoothness
thereby setting into motion the final clotting pro- and glycocalyx-thrombomodulin layer are lost, which
cess, as described earlier.! activates both factor XII and the platelets, thus setting off
the intrinsic pathway of clotting. If factor XII and platelets
Role of Calcium Ions in the Intrinsic and
come into contact with the subendothelial collagen, the
Extrinsic Pathways
activation is even more powerful.
Except for the first two steps in the intrinsic pathway, cal- Intact endothelial cells also produce other substances
cium ions are required for promotion or acceleration of such a prostacyclin and nitric oxide (NO) that inhibit
all the blood-clotting reactions. Therefore, in the absence platelet aggregation and initiation of blood clotting. Pros-
of calcium ions, blood clotting by either pathway does not tacyclin, also called prostaglandin I2 (PGI2), is a member
occur. of the eicosanoid family of lipids and is a vasodilator, as
In the living body, the calcium ion concentration well as an inhibitor of platelet aggregation. As discussed
seldom falls low enough to affect blood-clotting kinet- in Chapter 17, NO is a powerful vasodilator released from
ics significantly. However, when blood is removed from healthy vascular endothelial cells throughout the body,
someone, it can be prevented from clotting by reducing and it is an important inhibitor of platelet aggregation.
the calcium ion concentration below the threshold level When endothelial cells are damaged, their production of
for clotting by deionizing the calcium by causing it to prostacyclin and NO is greatly diminished.!
react with substances such as citrate ion or by precipitat-
ing the calcium with substances such as oxalate ion.! Antithrombin Action of Fibrin and Antithrombin III.
Among the most important anticoagulants in the blood
Interaction Between Extrinsic and are those that remove thrombin from the blood. The most
Intrinsic Pathways—Summary of Blood- powerful of these are the following: (1) the fibrin fibers
Clotting Initiation that are formed during the process of clotting; and (2)
It is clear from the schemas of the intrinsic and extrinsic an α globulin called antithrombin III or antithrombin-
systems that after blood vessels rupture, clotting occurs heparin cofactor.
by both pathways simultaneously. Tissue factor initiates While a clot is forming, about 85% to 90% of the throm-
the extrinsic pathway, whereas contact of factor XII and bin formed from the prothrombin becomes adsorbed to
platelets with collagen in the vascular wall initiates the the fibrin fibers as they develop. This adsorption helps
intrinsic pathway. prevent the spread of thrombin into the remaining blood
An especially important difference between the and, therefore, prevents excessive spread of the clot.
extrinsic and intrinsic pathways is that the extrin- The thrombin that does not adsorb to the fibrin
sic pathway can be explosive; once initiated, its speed fibers soon combines with antithrombin III. This further
of completion to the final clot is limited only by the blocks the effect of thrombin on the fibrinogen and then
amount of tissue factor released from the traumatized also inactivates thrombin itself during the next 12 to 20
tissues and by the quantities of factors X, VII, and V in minutes.!
483
UNIT VI Blood Cells, Immunity, and Blood Coagulation
tPA
Heparin. Heparin is another powerful anticoagulant but, converts plasminogen to plasmin, which in turn removes
because its concentration in the blood is normally low, the remaining unnecessary blood clot. In fact, many small
it has significant anticoagulant effects only under special blood vessels in which blood flow has been blocked by
physiological conditions. However, heparin is used widely clots are reopened by this mechanism. Thus, an especially
as a pharmacological agent in medical practice in much important function of the plasmin system is to remove
higher concentrations to prevent intravascular clotting. minute clots from millions of tiny peripheral vessels that
The heparin molecule is a highly negatively charged eventually would become occluded were there no way to
conjugated polysaccharide. By itself, it has little or no clear them.!
anticoagulant properties, but when it combines with
antithrombin III, the effectiveness of antithrombin III
for removing thrombin increases by a hundredfold to a CONDITIONS THAT CAUSE EXCESSIVE
thousandfold and thus acts as an anticoagulant. There- BLEEDING IN HUMANS
fore, in the presence of excess heparin, the removal of free Excessive bleeding can result from a deficiency of any of
thrombin from the circulating blood by antithrombin III the many blood-clotting factors. Three particular types of
is almost instantaneous. bleeding tendencies that have been studied to the greatest
The complex of heparin and antithrombin III removes extent are discussed here—bleeding caused by (1) vitamin
several other activated coagulation factors in addition to K deficiency, (2) hemophilia, and (3) thrombocytopenia
thrombin, further enhancing the effectiveness of antico- (platelet deficiency).
agulation. The others include activated factors IX through
XII. DECREASED PROTHROMBIN, FACTOR VII,
Heparin is produced by many different cells of the FACTOR IX, AND FACTOR X CAUSED BY
body, but the largest quantities are formed by the baso- VITAMIN K DEFICIENCY
philic mast cells located in the pericapillary connec-
tive tissue throughout the body. These cells continually With few exceptions, almost all the blood-clotting fac-
secrete small quantities of heparin that diffuse into the tors are formed by the liver. Therefore, diseases of the
circulatory system. The basophil cells of the blood, which liver such as hepatitis, cirrhosis, and acute yellow atrophy
are functionally almost identical to the mast cells, release (degeneration of the liver caused by toxins, infections, or
small quantities of heparin into the plasma. other agents) can sometimes depress the clotting system
Mast cells are abundant in tissue surrounding the cap- so much that the patient develops a severe tendency to
illaries of the lungs and, to a lesser extent, capillaries of the bleed.
liver. It is easy to understand why large quantities of hepa- Another cause of depressed formation of clotting
rin might be needed in these areas because the capillaries factors by the liver is vitamin K deficiency. Vitamin K
of the lungs and liver receive many embolic clots that have is an essential factor to a liver carboxylase that adds a
formed in slowly flowing venous blood; sufficient produc- carboxyl group to glutamic acid residues on five of the
tion of heparin prevents further growth of the clots.! important clotting factors—prothrombin, factor VII, fac-
tor IX, factor X, and protein C. On adding the carboxyl
PLASMIN CAUSES LYSIS OF BLOOD CLOTS group to glutamic acid residues on the immature clot-
ting factors, vitamin K is oxidized and becomes inactive.
The plasma proteins contain a euglobulin called plas-
Another enzyme, vitamin K epoxide reductase complex 1
minogen (profibrinolysin) that when activated, becomes a
(VKORC1), reduces vitamin K back to its active form. In
substance called plasmin (fibrinolysin). Plasmin is a pro-
the absence of active vitamin K, subsequent insufficiency
teolytic enzyme that resembles trypsin, the most impor-
of these coagulation factors in the blood can lead to seri-
tant proteolytic digestive enzyme of pancreatic secretion.
ous bleeding tendencies.
Plasmin digests fibrin fibers and some other protein
Vitamin K is continually synthesized in the intestinal
coagulants, such as fibrinogen, factor V, factor VIII, pro-
tract by bacteria, so vitamin K deficiency seldom occurs
thrombin, and factor XII. Therefore, whenever plasmin
in healthy persons as a result of the absence of vitamin K
is formed, it can cause lysis of a clot by destroying many
from the diet (except in neonates, before they establish
of the clotting factors, thereby sometimes even causing
their intestinal bacterial flora). However, in persons with
hypocoagulability of the blood.
gastrointestinal disease, vitamin K deficiency often occurs
Activation of Plasminogen to Form Plasmin, Then as a result of poor absorption of fats from the gastrointes-
Clot Lysis. When a clot is formed, a large amount of plas- tinal tract because vitamin K is fat-soluble and is ordinar-
minogen is trapped in the clot, along with other plasma ily absorbed into the blood along with the fats.
proteins. This will not become plasmin or cause lysis of One of the most prevalent causes of vitamin K defi-
the clot until it is activated. The injured tissues and vascu- ciency is failure of the liver to secrete bile into the gas-
lar endothelium very slowly release a powerful activator trointestinal tract, which occurs as a result of obstruction
called tissue plasminogen activator (t-PA); a few days later, of the bile ducts or of liver disease. Lack of bile prevents
after the clot has stopped the bleeding, t-PA eventually adequate fat digestion and absorption and, therefore,
484
Chapter 37 Hemostasis and Blood Coagulation
depresses vitamin K absorption as well. Thus, liver dis- disease with somewhat different characteristics, called
ease often causes decreased production of prothrombin von Willebrand disease, results from loss of the large
and some other clotting factors because of poor vitamin component.
K absorption and because of the diseased liver cells. As When a person with classic hemophilia experiences
a result, vitamin K is injected into surgical patients with severe prolonged bleeding, almost the only therapy that is
UNIT VI
liver disease or with obstructed bile ducts before the truly effective is injection of purified factor VIII or factor
surgical procedure is performed. Ordinarily, if vitamin IX. Both these clotting factors are now available as recom-
K is given to a deficient patient 4 to 8 hours before the binant proteins, although they are expensive and their
operation and the liver parenchymal cells are at least half- half-lives are relatively short; therefore, these products are
normal in function, sufficient clotting factors will be pro- not readily available for many patients with hemophilia,
duced to prevent excessive bleeding during the operation.! especially in economically disadvantaged countries.!
HEMOPHILIA THROMBOCYTOPENIA
Hemophilia is a bleeding disease that occurs almost Thrombocytopenia means the presence of very low num-
exclusively in males. In 85% of cases, it is caused by an bers of platelets in the circulating blood. People with
abnormality or deficiency of factor VIII; this type of hemo- thrombocytopenia have a tendency to bleed, as do hemo-
philia is called hemophilia A or classic hemophilia. About philiacs, except that the bleeding is usually from many
1 of every 10,000 males in the United States has classic small venules or capillaries, rather than from larger ves-
hemophilia. In the other 15% of patients with hemophilia sels, as in hemophilia. As a result, small punctate hem-
B, the bleeding tendency is caused by deficiency of factor orrhages occur throughout all the body tissues. The skin
IX. Both these factors are transmitted genetically by way of such a person displays many small petechiae, red or
of the female (X) chromosome and are recessive in their purplish blotches, giving the disease the name throm-
inheritance. Therefore, a woman will rarely have hemo- bocytopenic purpura. As noted, platelets are especially
philia because at least one of her two X chromosomes will important for the repair of minute breaks in capillaries
have the appropriate genes. If one of her X chromosomes and other small vessels.
is deficient, she will be a hemophilia carrier; her male off- Platelet counts below 30,000/µl, compared with the
spring will have a 50% chance of inheriting the illness, and normal value of 150,000 to 450,000/µl, increase the risk
her female offspring will have a 50% chance of inheriting for excessive bleeding after surgery or injury. Spontane-
the carrier status. ous bleeding, however, will not ordinarily occur until the
Although female carriers have one normal allele and number of platelets in the blood falls below 30,000/µl.
usually do not develop symptomatic hemophilia, some Levels as low as 10,000/µl are frequently lethal.
may experience a mild bleeding trait. It is also possible Even without determining specific platelet counts in
for female carriers to develop mild hemophilia due to the blood, sometimes one can suspect the existence of
loss of part or all of the normal X chromosome (as in thrombocytopenia if the person’s blood clot fails to retract.
Turner syndrome) or inactivation (lyonization) of the As noted earlier, clot retraction is normally dependent
X-chromosomes. For a female to inherit full-blown on release of multiple coagulation factors from the large
symptomatic hemophilia A or B, she must receive two numbers of platelets entrapped in the fibrin mesh of the
deficient X-chromosomes, one from her carrier mother clot.
and the other from her father, who must have hemophilia. The major causes of thrombocytopenia include the
Most cases of hemophilia are inherited, but approxi- following: (1) decreased platelet production in the bone
mately one-third of hemophilia patients do not have a marrow due to infections or sepsis, nutrient deficiencies,
family history of the disease, which appears to be caused or myelodysplastic disorders, which usually also reduce
by novel mutation events. production of other cells (red blood cells [RBCs] and
The bleeding trait in hemophilia can have various white blood cells); (2) peripheral platelet destruction by
degrees of severity, depending on the genetic deficiency. antibodies; (3) sequestration (pooling) of platelets in the
Bleeding usually does not occur except after trauma, but spleen, especially in individuals with portal hypertension
in some patients, the degree of trauma required to cause and excessively large spleens (splenomegaly); (4) con-
severe and prolonged bleeding may be so mild that it is sumption of platelets in thrombi; and (4) dilution of the
hardly noticeable. For example, bleeding can often last for blood from fluid resuscitation or massive transfusion.
days after extraction of a tooth. Most people with thrombocytopenia have the disease
Factor VIII has two active components, a large compo- known as idiopathic thrombocytopenia, which means
nent with a molecular weight in the millions and a smaller thrombocytopenia of unknown cause. In most of these
component with a molecular weight of about 230,000. people, it has been discovered that, for unknown reasons,
The smaller component is most important in the intrinsic specific antibodies have formed and react against the plate-
pathway for clotting, and it is deficiency of this part of fac- lets to destroy them. Relief from bleeding for 1 to 4 days
tor VIII that causes classic hemophilia. Another bleeding can often be effected in a patient with thrombocytopenia
485
UNIT VI Blood Cells, Immunity, and Blood Coagulation
by giving fresh whole blood transfusions that contain large artery is blocked, death may not occur, or the embolism
numbers of platelets. Also, splenectomy may be helpful, may lead to death a few hours to several days later because
sometimes resulting in an almost complete cure because of further growth of the clot in the pulmonary vessels.
the spleen normally removes large numbers of platelets However, again, t-PA therapy can be a lifesaver.!
from the blood.!
DISSEMINATED INTRAVASCULAR
COAGULATION
THROMBOEMBOLIC CONDITIONS
Occasionally, the clotting mechanism becomes activated
Thrombi and Emboli. An abnormal clot that develops in widespread areas of the circulation, giving rise to the
in a blood vessel is called a thrombus. Once a clot has de- condition called disseminated intravascular coagulation
veloped, continued flow of blood past the clot is likely to (DIC). This condition often results from the presence
break it away from its attachment and cause the clot to of large amounts of traumatized or dying tissue in the
flow with the blood; such freely flowing clots are known body that releases great quantities of tissue factor into
as emboli. Also, emboli that originate in large arteries or the blood. Frequently, the clots are small but numerous,
in the left side of the heart can flow peripherally and plug and they plug a large share of the small peripheral blood
arteries or arterioles in the brain, kidneys, or elsewhere. vessels. This process occurs especially in patients with
Emboli that originate in the venous system or in the right widespread septicemia, in which circulating bacteria or
side of the heart generally flow into the lungs to cause pul- bacterial toxins—especially endotoxins—activate the clot-
monary arterial embolism.! ting mechanisms. The plugging of small peripheral vessels
greatly diminishes delivery of oxygen and other nutrients
Causes of Thromboembolic Conditions. The causes of to the tissues, a situation that leads to or exacerbates cir-
thromboembolic conditions in people are usually twofold: culatory shock. It is partly for this reason that septicemic
(1) a roughened endothelial surface of a vessel—as may be shock is lethal in 35% to 50% of patients.
caused by arteriosclerosis, infection, or trauma—is likely A peculiar effect of disseminated intravascular coagu-
to initiate the clotting process; and (2) blood often clots lation is that the patient, on occasion, begins to bleed. The
when it flows very slowly through blood vessels, where reason for this bleeding is that so many of the clotting fac-
small quantities of thrombin and other procoagulants are tors are removed by the widespread clotting that too few
always being formed.! procoagulants remain to allow normal hemostasis of the
Use of Tissue Plasminogen Activator in Treating In- remaining blood.!
travascular Clots. Genetically engineered tissue plas-
minogen activator (t-PA) is available. When delivered ANTICOAGULANTS FOR CLINICAL USE
through a catheter to an area with a thrombus, it is effec-
tive in activating plasminogen to plasmin, which in turn In some thromboembolic conditions, it is desirable to
can dissolve some intravascular clots. For example, if used delay the coagulation process. Various anticoagulants
within the 1 or 2 hours after thrombotic occlusion of a have been developed for this purpose. The ones most
coronary artery, the heart is often spared serious damage.! clinically useful are heparin and the coumarins.
486
Chapter 37 Hemostasis and Blood Coagulation
discussed previously, this enzyme converts the inactive, polymerized into glucose or metabolized directly for
oxidized form of vitamin K to its active, reduced form. energy. Consequently, 500 milliliters of blood that has
By inhibiting VKORC1, warfarin decreases the avail- been rendered noncoagulable by citrate can ordinarily be
able active form of vitamin K in the tissues. When this transfused into a recipient within a few minutes, without
decrease occurs, the coagulation factors are no longer car- dire consequences. However, if the liver is damaged, or
UNIT VI
boxylated and are biologically inactive. Over several days, if large quantities of citrated blood or plasma are given
the body stores of the active coagulation factors degrade too rapidly (within fractions of a minute), the citrate ion
and are replaced by inactive factors. Although the coagu- may not be removed quickly enough, and the citrate can,
lation factors continue to be produced, they have greatly under these conditions, greatly depress the level of cal-
decreased coagulant activity. cium ion in the blood, which can result in tetany and con-
After administration of an effective dose of warfarin, vulsive death.!
the coagulant activity of the blood decreases to about 50%
of normal by the end of 12 hours and to about 20% of
BLOOD COAGULATION TESTS
normal by the end of 24 hours. In other words, the coagu-
lation process is not blocked immediately but must await
BLEEDING TIME
the degradation of the active prothrombin and the other
affected coagulation factors already present in the plasma. When a sharp-pointed knife is used to pierce the tip of the
Normal coagulation usually returns 1 to 3 days after dis- finger or earlobe, bleeding ordinarily lasts for 1 to 6 min-
continuing coumarin therapy.! utes. This time depends largely on the depth of the wound
and degree of hyperemia in the finger or earlobe at the
PREVENTION OF BLOOD COAGULATION time of the test. Lack of any one of several of the clotting
OUTSIDE THE BODY factors can prolong the bleeding time, but it is especially
prolonged by lack of platelets.!
Although blood removed from the body and held in a
glass test tube normally clots in about 6 minutes, blood CLOTTING TIME
collected in siliconized containers often does not clot for
Many methods have been devised for determining blood-
1 hour or more. The reason for this delay is that preparing
clotting time. The one most widely used is to collect
the surfaces of the containers with silicone prevents con-
blood in a chemically clean glass test tube and then to
tact activation of platelets and factor XII, the two princi-
tip the tube back and forth about every 30 seconds until
pal factors that initiate the intrinsic clotting mechanism.
the blood has clotted. By this method, the normal clot-
Conversely, untreated glass containers allow contact acti-
ting time is 6 to 10 minutes. Procedures using multiple
vation of the platelets and factor XII, with the rapid devel-
test tubes have also been devised for determining clotting
opment of clots.
time more accurately.
Heparin can be used for preventing coagulation of
Unfortunately, the clotting time varies widely, depend-
blood outside the body, as well as in the body. Heparin is
ing on the method used for measuring it, so it is no lon-
especially used in surgical procedures in which the blood
ger used in many clinics. Instead, measurements of the
must be passed through a heart-lung machine or artificial
clotting factors themselves are made, using sophisticated
kidney machine and then back into the patient.
Various substances that decrease the concentration of chemical procedures.!
calcium ions in the blood can also be used for preventing PROTHROMBIN TIME AND
blood coagulation outside the body. For example, a solu- INTERNATIONAL NORMALIZED RATIO
ble oxalate compound mixed in a very small quantity with
a sample of blood causes precipitation of calcium oxalate The prothrombin time indicates the concentration of
from the plasma and thereby decreases the ionic calcium prothrombin in the blood. Figure 37-7 shows the rela-
level so much that blood coagulation is blocked. tionship of prothrombin concentration to prothrombin
Any substance that deionizes the blood calcium will time. The method for determining prothrombin time is
prevent coagulation. The negatively charged citrate ion is the following.
especially valuable for this purpose; it is mixed with blood Blood removed from the patient is immediately oxa-
usually in the form of sodium, ammonium, or potassium lated so that none of the prothrombin can change into
citrate. The citrate ion combines with calcium in the thrombin. Then, a large excess of calcium ion and tis-
blood to produce a nonionized calcium compound, and sue factor is quickly mixed with the oxalated blood. The
the lack of ionic calcium prevents coagulation. Citrate excess calcium nullifies the effect of the oxalate, and the
anticoagulants have an important advantage over the oxa- tissue factor activates the prothrombin to thrombin reac-
late anticoagulants because oxalate is toxic to the body, tion by means of the extrinsic clotting pathway. The time
whereas moderate quantities of citrate can be injected required for coagulation to take place is known as the
intravenously. After injection, the citrate ion is removed prothrombin time. The shortness of the time is determined
from the blood within a few minutes by the liver and is mainly by the prothrombin concentration. The normal
487
UNIT VI Blood Cells, Immunity, and Blood Coagulation
100 Tests similar to that for prothrombin time and INR have
been devised to determine the quantities of other blood-
clotting factors. In each of these tests, excesses of calcium
Concentration (percent of normal)
ions and all the other factors in addition to the one being
tested are added to oxalated blood all at once. Then, the time
required for coagulation is determined in the same manner
as for prothrombin time. If the factor being tested is defi-
50.0 cient, the coagulation time is prolonged. The time itself can
then be used to quantitate the concentration of the factor.
25.0 Bibliography
12.5 Becker RC, Sexton T, Smyth SS: Translational implications of platelets
6.25 as vascular first responders. Circ Res 122:506, 2018.
0 Furie B, Furie BC: Mechanisms of thrombus formation. N Engl J Med
0 10 20 30 40 50 60 359:938, 2008.
Gupta S, Shapiro AD: Optimizing bleed prevention throughout the
Prothrombin time
(seconds) lifespan: womb to tomb. Haemophilia 24 Suppl 6:76, 2018.
Hess CN, Hiatt WR: Antithrombotic therapy for peripheral artery dis-
Figure 37-7. Relationship of prothrombin concentration in the blood ease in 2018. JAMA 319:2329, 2018.
to prothrombin time. Hunt BJ: Bleeding and coagulopathies in critical care. N Engl J Med
370:847, 2014.
Koupenova M, Clancy L, Corkrey HA, Freedman JE: Circulating plate-
prothrombin time is about 12 seconds. In each laboratory, lets as mediators of immunity, inflammation, and thrombosis. Circ
a curve relating prothrombin concentration to prothrom- Res 122:337, 2018.
bin time, such as that shown in Figure 37-7, is drawn for Kucher N: Clinical practice. Deep-vein thrombosis of the upper ex-
the method used so that the prothrombin in the blood can tremities. N Engl J Med 364:861, 2011.
Leebeek FW, Eikenboom JC: Von Willebrand’s disease. N Engl J Med
be quantified. 375:2067, 2016.
The results obtained for prothrombin time may vary Luyendyk JP, Schoenecker JG, Flick MJ: The multifaceted role of fi-
considerably, even in the same individual if there are brinogen in tissue injury and inflammation. Blood 133:511, 2019.
differences in activity of the tissue factor and the ana- Maas C, Renné T: Coagulation factor XII in thrombosis and inflamma-
lytical system used to perform the test. Tissue factor tion. Blood 131:1903, 2018.
McFadyen JD, Schaff M, Peter K: Current and future antiplatelet
is isolated from human tissues, such as placental tis- therapies: emphasis on preserving haemostasis. Nat Rev Cardiol
sue, and different batches may have different activity. 15:181, 2018.
The international normalized ratio (INR) was devised Mohammed BM, Matafonov A, Ivanov I, et al: An update on factor XI
as a way to standardize measurements of prothrombin structure and function. Thromb Res 161:94, 2018.
time. For each batch of tissue factor, the manufacturer Nachman RL, Rafii S: Platelets, petechiae, and preservation of the vas-
cular wall. N Engl J Med 359:1261, 2008.
assigns an international sensitivity index (ISI), which Negrier C, Shima M, Hoffman M: The central role of thrombin in bleed-
indicates the activity of the tissue factor with a stan- ing disorders. Blood Rev 2019 May 22. pii: S0268-960X(18)30097-3.
dardized sample. The ISI usually varies between 1.0 https://www.doi.org/10.1016/j.blre.2019.05.006
and 2.0. The INR is the ratio of the person’s prothrom- Peters R, Harris T: Advances and innovations in haemophilia treat-
bin time (PT) to a normal control sample raised to the ment. Nat Rev Drug Discov 17:493, 2018.
Samuelson Bannow B, Recht M, Négrier C, et al: Factor VIII: long-
power of the ISI: established role in haemophilia A and emerging evidence beyond
ISI haemostasis. Blood Rev 35:43, 2019.
INR = PTtest
Tillman BF, Gruber A, McCarty OJT, Gailani D: Plasma contact factors
PT normal
as therapeutic targets. Blood Rev 32:433, 2018.
The normal range for INR in a healthy person is 0.9 to van der Meijden PEJ, Heemskerk JWM: Platelet biology and functions:
1.3. A high INR level (e.g., 4 or 5) indicates a high risk of new concepts and clinical perspectives. Nat Rev Cardiol 16:166, 2019.
Wells PS, Forgie MA, Rodger MA: Treatment of venous thromboem-
bleeding, whereas a low INR (e.g., 0.5) suggests that there bolism. JAMA 311:717, 2014.
is a chance of having a clot. Patients undergoing warfarin Weyand AC, Pipe SW: New therapies for hemophilia. Blood 133:389,
therapy usually have an INR of 2.0 to 3.0. 2019.
488
Lymphoreticular system
Fourteenth Edition
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Intended Learning Outcomes
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Early Transfusions
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Agglutinins
Antibodies, mostly
IgM and IgG
Begin developing
age 2 8 months,
peak ~age 10 years
Response to A and
B antigens in foods,
bacteria; initial
exposures are
environmental
Figure 36-1
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Blood Groups
Table 36-1
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Transfusion Reactions
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
The Rh (rhesus) Antigens
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Anti-Rh Transfusion Reactions (1 of 2)
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Agglutinins
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Rh Blood Types
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Universal Donor; Suitable for all?
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Blood Transfusion
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Hemolytic Disease of the Newborn
(Erythroblastosis Fetalis)
ABO incompatibility
O mother and A or B fetus
Most anti-A is IgM which does not cross placenta
ABO antigens are not well developed in fetus.
Rh incompatibility
Rh (D) positive fetus and Rh negative mother
Immunization due to fetal-maternal bleeding during delivery. Mother develops Anti-
D agglutinins
Usually not a problem with first pregnancy
Worse with subsequent pregnancies (3% EF second pregnancy, 10% with third)
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Hemolytic Disease of the Newborn-
Overview
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Hemolytic Disease of the Newborn
(Erythroblastosis Fetalis) Pathophysiology
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Hemolytic Disease of the Newborn
(Erythroblastosis Fetalis) Treatment
Hemolytic anemia:
If severe: treated with exchange
transfusion: Replace baby blood with
Rh- ve RBC (several times)
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Hemolytic Disease of the Newborn
(Erythroblastosis Fetalis) Prevention
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Blood Transfusion
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Transfusion Reactions
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Acute Renal Failure After
Transfusion Reaction
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Graft Acceptance/Rejection
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Rejection
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Avoidance or Suppression of
Rejection
Tissue typing
Blood type
HLA (MHC) antigens
Immunosuppressive drugs
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
HLA Antigens
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Histocompatibility Testing
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Immunosuppressive Drugs
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Thank you
Dr. Reham Khalaf-Nazzal, MD, PhD Copyright © 2021 by Saunders, an imprint of Elsevier Inc.
Pathology and microbiology
Regulation:
Primarily EPO (Endothelial cells in the peritubular capillaries of the Kidneys and
hepatocytes).
Stages:
Hematopoietic stem cell (erythroid progenitor cell):
Proerythroblast
In bone Erythroblast
marrow Normoblast.
Reticulocyte:
acidophilic cytoplasm with granules composed of cytoplasmic
libosomal RNA (enabling reticulocytes to be stained with methylene
blue)
Reticulocytes are released into the blood.
The peripheral reticulocyte count reflects erythropoietic activity.
In Increased reticulocytes indicate increased erythropoiesis (e.g.
blood due to hemolysis).
Decreased reticulocytes indicate decreased erythropoiesis (e.g.,
due to aplastic anemia).
Reticulocytes mature into erythrocytes.
MCV vs MCH
Mean cell volume (MCV): Mean cell hemoglobin (MCH):
The average volume per red cell, the average mass of hemoglobin per
expressed in femtoliters (𝝁𝒎𝟑 ). red cell, expressed in picograms
A) Anemia
Decrease in hemoglobin concentration. RBC count, and/or hematocrit.
General principles:
1) Reduction in circulating red blood cell (RBC) mass.
2) Presents with signs and symptoms of hypoxia (general features of anemia):
Weakness, fatigue, and dyspnea.
Pale conjunctiva and skin.
Headache and light headedness.
Angina, especially with preexisting coronary artery disease.
Dizziness, difficulty of concentration and insomnia.
3) Hemoglobin (Hb), hematocrit (Hct), and RBC count are used as surrogates for
RBC mass, which is difficult to measure.
Men: < 𝟏𝟑. 𝟓 𝒈/𝒅𝒍
Women: < 𝟏𝟐 𝒈/𝒅𝒍
Children: 6 - 59 months < 𝟏𝟏 𝒈/𝒅𝒍
The fetus may reach 20-22 within 30 5 -11 years < 𝟏𝟏. 𝟓 𝒈/𝒅𝒍
days. 12 -14 years < 𝟏𝟐 𝒈/𝒅𝒍
Grading of anemia:
Grades Hemoglobin count
Normal 𝒉𝒊𝒈𝒉𝒆𝒓 𝒐𝒓 𝒆𝒒𝒖𝒂𝒍 − 𝟏𝟏
Mild 𝟗. 𝟓 – 𝟏𝟏
Moderate 𝟖 – 𝟗. 𝟓
Serious or sever 𝟔. – 𝟖
Life threatening 𝒍𝒆𝒔𝒔 𝒕𝒉𝒂𝒏 − 𝟔
Classification of anemia:
Anemia may be classified into several subtypes based on the following methods:
Morphology/size of RBCs (the classification most widely used).
Time course: acute vs. chronic
Inheritance: inherited vs. acquired
Etiology: primary vs. secondary.
RBC proliferation: hypoproliferative (decreased RBC production) vs.
hyperproliferative (increased RBC destruction or blood loss).
MICROCYTIC ANEMIAS
A. Anemia with MCV < 𝟖𝟎 𝝁𝒎𝟑 .
B. Microcytic anemias are due to decreased production of hemoglobin.
RBC progenitor cells in the bone marrow are large and normally divide
multiple times to produce smaller mature cells (MCV = 80-100 µm3).
Microcytosis is due to an "extra" division which occurs to maintain
hemoglobin concentration.
C. A decrease in heme, globin, iron or protoporphyrin. leads to microcytic
anemia.
D. Microcytic anemias include;
Epidemiology
Children up to 5 years of age.
Young women of child-bearing age (due to menstrual blood loss).
Pregnant women
2) Decreased intake:
Chronic undernutrition.
Cereal-based diet ()النظام الغذائي القائم على الحبوب.
4) Increased demand
Pregnancy.
Growth spurt ()النمو السريع جدا.
Erythropoietin (EPO) therapy.
Clinical features:
Fatigue, lethargy.
Signs and
symptoms of Pallor (primarily seen in highly vascularized mucosa, e.g., the
anemia: conjunctiva).
Treatment:
1) Dietary modifications:
All patients:
Encourage consumption of iron-rich foods.
prevent patients from taking non supplements to avoid the following substances
that reduce non absorption:
Food: tea, cereals, daily products
Drugs: calcium, antacids, protein pump inhibitors {PPIs}
Etiology
Inflammation (e.g., rheumatoid arthritis, systemic lupus erythematosus).
Malignancy.
Chronic infections.
Diagnostics:
Normocytic anemia (early phase) microcytic anemia (late phase).
(↓) available iron → (↓) heme → (↓) hemoglobin → microcytic anemia.
Treatment:
Treat the underlying cause.
Blood transfusion if required.
EPO in chronic incurable diseases (e.g., chronic kidney disease).
3) SIDEROBLASTIC ANEMIA
Defective heme metabolism, which leads to iron trapping inside
the mitochondria.
4) THALASSEMIA
Genetic disorders resulting from decreased biosynthesis of globin chains of
hemoglobin.
Due to mutations in and around the globin genes.
Decreased production of one or more of the globin chains.
Result in an imbalance in the relative amounts of the 𝛂- and non 𝛂-chains.
Altered α /non- α ratio.
As a consequences of thalassemia there is excess production of the other chains,
and a decreased over-all hemoglobin synthesis.
Thalassemia provides partial resistance against malaria.
Epidemiology:
Beta thalassemia: most commonly seen in people of Mediterranean descent
Alpha thalassemia: most commonly seen in people of Asian and African descent
Etiology:
General cause; gene mutations.
Pathophysiology:
Increased hemolysis: erythrocyte instability with hemolysis.
Anemia → ↑ erythropoietin → bone marrow hyperplasia and skeletal
deformities.
Beta thalassemia:
In a normal cell: The (𝜷-globin chains are coded by a total
of two alleles.
Beta thalassemia minor (trait): one defective allele
Beta thalassemia major (Cooley Two defective alleles.
anemia):
A combination of one defective (𝜷 -globin
Sickle cell beta thalassemia: allele and one defective HbS allele.
Unremarkable symptoms.
2) Major variant:
A) CBC:
Microcytic hypochromic anemia.
B) Hemolysis evaluation:
Nonimmune-mediated hemolytic anemia.
↓ Haptoglobin, ↑ LDH ↑ reticulocytes. HALT:
Hyperbilirubinemia (indirect). H: Hemoglobin H
C) Peripheral blood smear findings include: A: Asplenia
Target cells. L: Liver diseases
Teardrop cells (highly in beta thalassemia).
Anisopoikilocytosis. T: Thalassemia
Low ferritin suggests Iron deficiency anemia and patients should receive iron
supplementation.
Suspect thalassemia if there is no significant response after three months.
Complications of thalassemia:
A. Iron overload disease:
o All patients receiving transfusion therapy should be periodically
evaluated for non-overload disease and subsequent organ damage.
B. Hepatobiliary complications Cholelithiasis
C. Hematologic complications Hypercoagulable states Hemolytic crisis.
D. Extramedullary hematopoietic pseudotumors.
E. Cardiovascular complications.
F. Chronic leg ulcers.
G. Mental health complications.
Management of thalassemia:
Thalassemia minor:
2) Folate deficiency:
3) Fanconi anemia
Fanconi anemia is a rare disease passed down through families (inherited), that
mainly affects the bone marrow.
It results in decreased production of all types of blood cells. This is the most
common inherited form of aplastic anemia.
4) Orotic aciduria
III. Drugs:
Metabolic inhibitors.
Inborn errors.
Unexplained disorders.
Folate and vitamin B12 are necessary for synthesis of DNA precursors.
A. Folate circulates in the serum as methyltetrahydrofolate (methyl THF); removal of
the methyl group allows for participation in the synthesis of DNA precursors.
B. Methyl group is transferred to vitamin B12 (cobalamin).
C. Vitamin B12 then transfers it to homocysteine, producing methionine.
Clinical features for anemia in general:
1) Asymptomatic:
2) One or more of the following symptoms.
Pallor (e.g., on mucous membranes, conjuiictivae).
Exertional dyspnea and fatigue.
Pica (craving for ice or dirt).
Jaundice (in hemolytic anemia).
Muscle cramps.
Growth impairment (chronic anemia).
Worsening of angina pectoris.
Features of hyperdynamic state.
Bounding pulses ()االحساس بنبض قوي.
Tachycardia/palpitatioiis.
Flow murmur.
Pulsatile sound in the ear.
Possibly heart failure (anemia-induced heart failure).
Features of extramedullary hematopoiesis may be present in certain
severe, chronic forms of anemia (e.g., thalassemia, myelofibrosis).
Hepatosplenomegaly.
Paravertebral mass.
Widening of diploic spaces of the skull.
7- Biopsy:
Aplastic anemia
Damage to hematopoietic stem cells, resulting in pancytopenia (anemia,
thrombocytopenia, and leukopenia) with low reticulocyte count.
Should not be confused with aplastic crisis, a condition in which erythropoiesis
is temporarily suppressed (e.g., due to parvovirus B19 infection hi patients with
hemolytic anemias).
Etiologies:
Idiopathic > 𝟓𝟎% of cases [Possibly immune-mediated]
Medication side Carbamazepine, metliimazole. NSAIDs, chloramphenicol.,
effects: propylthiouracil sulfa drugs, cytostatic drugs (esp.
alkylating agents and antimetabolites)
Toxins: benzene
Viruses: HBV, EBV, CMV and HIV
Fanconi anemia This is the most common inherited form of aplastic anemia.
Ionizing
radiation
Clinical features:
Fatigue, malaise.
Pallor.
Infection.
Purpura, petechiae, mucosal bleeding.
1) CBC:
Pancytopenia.
Normocytic or macrocytic anemia (funconi anemia)
Reticulocytes EPO
Low High
Treatment:
1) Cessation of the causative agent.
2) Supportive therapy: Myelophthisic anemia is a
Treatment of infections. normocytic, normochromic anemia
that occurs when normal marrow
Blood transfusion.
space is infiltrated and replaced by
Platelet transfusion.
nonhematopoietic or abnormal cells.
3) Bone marrow stimulants: Causes include tumors, granulomatous
disorders, lipid storage diseases, and
GM-CSF. primary myelofibrosis.
4) Immunosuppressive therapy:
Cyclosporine, Antithymocyte globulin (ATG), hematopoietic cell transplantation
(HCT) in young patients.
Basophilic stippling
Distinctive property of pure
Pure red cell aplasia:
Normocytic, normochromic anemia characterized by a
severe reduction in circulating reticulocytes and marked
reduction or absence of erythroid precursors in the bone
marrow.
The major difference between PRCA and aplastic anemia is that, in PRCA, only the red
blood cell line is affected, while the white blood cells and platelets remain at normal
levels. In aplastic anemia, all three blood cell types are typically affected.
Hemolytic Anemia
The term given to a large group of anemias that are caused by the premature
destruction / hemolysis of circulating red blood cells (RBCs).
Epidemiology:
5% of all cases of anemia.
Hereditary causes present early in life.
Autoimmune hemolytic anemia (AIHA) is more common in
middle-aged and older adults.
Males are predominantly affected by X-linked hereditary
spherocytosis.
Classification:
According to the cause of hemolysis: According the location of hemolysis:
intrinsic or extrinsic Intravascular or extravascular
Pathophysiology
1) Extravascular hemolysis:
Normal breakdown of RBCs occurs in the spleen every 120 days.
2) Intravascular hemolysis:
RBCs become fragmented when passing through narrowed vessel lumen
(microangiopathic) or prosthetic valves (macroangiopathic).
Clinical features
Jaundice.
Pigmented gallstones.
1) Signs of hemolysis Splenomegaly.
Back pain and dark urine in severe
hemolysis with hemoglobinuria.
Extravascular hemolysis
Direct Coombs test (positive for IgM / IgG).
Blood film: spherocytes, sickle cell anemia,
agglutination (in IgM Autoimmune hemolytic anemia
{AIHA}).
IgG binds RBCs in the relatively warm temperature of the central body
(warm agglutinin); membrane of antibody-coated RBC is consumed by splenic
macrophages, resulting in spherocytes.
Associated with SLE (most common cause), chronic lymphocytic
leukemia {CLL}, and certain drugs (classically, penicillin and cephalosporins).
Drug may attach to RBC membrane (e.g., penicillin) with subsequent
binding of antibody to drug-membrane complex.
Drug may induce production of autoantibodies (e.g., α-methyldopa)
that bind self-antigens on RBCs.
Treatment involves cessation of the offending drug, steroids, Intravenous
immune globulin (IVIG), and, if necessary, splenectomy.
1. IgM binds RBCs and fixes complement in the relatively cold temperature of
the extremities (cold agglutinin).
2. RBCs inactivate complement, but residual C3b serves as an opsonin for
splenic macrophages resulting in spherocytes; extreme activation of
complement can lead to intravascular hemolysis.
3. Associated with Mycoplasma pneumoniae and infectious mononucleosis.
4. Maybe found reticulocytes.
D) Coombs test is used to diagnose IHA; testing can be direct or indirect.
Direct Coombs test Indirect Coombs test
Transfusion medicine
Clinical Workup of autoimmune (workup and prevention of
applications hemolytic anemia transfusion reactions).
Perinatal care (workup and
prevention of HDFN).
Location of
antibodies RBC surface Serum (free)
detected
Coombs serum The patient's purified The patient's purified serum
added to: RESCs which has been mixed with
test RBCs.
AHGs in Antibodies and/or The patient's antibodies which
Coombs serum complement already are bound to test RBCs.
bind to: coating the patient's RBCs.
When anti-IgG/complement
is added to patient RBCs,
confirms the presence of agglutination occurs if RBCs
Direct Coombs antibody- or complement- are already coated with IgG or
test coated RBCs. complement.
This is the most important
test for IHA.
Anti IgG and test RBCs are
Indirect Coombs Confirms the presence of mixed with the patient serum;
test antibodies in patient serum. agglutination occurs if serum
antibodies are present.
Kidney disease.
Etiology:
Primary Thrombotic thrombocytopenic purpura.
MAHA Hemolytic uremic syndrome.
Autoimmune disease (e.g., SLE).
Hemolysis, Elevated Liver enzymes and Low Platelets {HELLP} syndrome.
Secondary
Hypertensive emergency.
MAHA
Disseminated intravascular coagulation (DIC).
Drug induced (e.g., quinine, trimethoprim /sulfamethoxazole, cyclosporine).
Pathophysiology:
Systemic microthrombi plug small vessels physical intravascular shearing
of RBCs that pass through the small vessels intravascular hemolysis,
schistocytes, and (↑) free Hb.
Clinical features:
Features of anemia.
Organ dysfunction due to microthrombi formation (e.g., renal dysfunction,
altered mental status).
Pathophysiology:
RBC destruction in the systemic circulation (large vessels) due to mechanical forces
applied to RBC membrane intravascular hemolysis, schistocytes, ↑ free Hb.
Etiology:
Autosomal recessive defect of pyruvate kinase.
Pathophysiology:
Glucose is the only energy source in RBCs.
Pyruvate kinase catalyzes the last step of glycolysis (i.e., irreversibly converts
phosphoenolpyruvate into pyruvate).
Absence of pyruvate kinase → ATP deficiency in RBC.
ATP deficiency disrupts the cation gradient along the RBC membrane rigid
RBCs (↑) hemolysis (extravascular - tissues).
Accumulation of 2,3-bisphosphoglycerate (↑) release of O2 from Hb masks
symptoms of anemia.
Clinical symptoms:
Usually asymptomatic.
Typically newborn jaundice due to hemolysis and history of exchange transfusions.
Splenomegaly.
Pallor, fatigue, weakness.
In rare cases: hydrops fetalis. Severe anemia the child may die
during birth or a few days after birth.
Diagnosis of pyruvate kinase deficiency:
(↓) Pyruvate kinase enzyme activity, PKLR gene mutation, Blood smear: burr
cells.
Hemoglobin Zurich
Pathophysiology:
Replacement of distal histidine in the beta-globin
chain with arginine enlargement of the ligand-
binding space around iron increased affinity for
carbon monoxide increased carboxyhemoglobin
levels (≥ 3%) Oxidative stress Formation of
Heinz body and hemolysis.
Normal level of
carboxyhemoglobin is 1-2 %
Hemoglobin C disease
Hemoglobin Occurs in individuals who are homozygous for the hemoglobin C
C disease mutation (HbCC).
Hemoglobin Occurs in individuals who are heterozygous carriers of the
C trait hemoglobin C mutation (HbAC).
Pathophysiology:
β-globin mutation (glutamate replaced by lysine).
HbC precipitates as crystals (↑) RBC rigidity and
(↓) deformability extravascular hemolysis.
HbC is less soluble than HbA and tends to form
hexagonal crystals, which lead to RBC dehydration (↑)
MCHC).
RBCs have reduced oxygen-binding capacity and a shorter lifespan.
Glucose-6-phosphate dehydrogenase deficiency (Favism)
Epidemiology:
G6PD deficiency is the most common human enzyme deficiency.
Prevalence: ∼ 400 million worldwide.
Affects primarily males of African, Mediterranean, and Asian descent.
Pathophysiology:
X-linked recessive inheritance.
G6PD is the rate-limiting enzyme of the pentose phosphate pathway (also
known as the hexose monophosphate shunt).
This pathway yields NADPH, which is essential for converting oxidized
glutathione back to its reduced form.
Reduced glutathione is capable of neutralizing reactive oxygen species
(ROS) and free radicals and therefore protecting RBCs from oxidative
damage.
In the absence of reduced glutathione (e.g., due to G6PD deficiency), RBCs
become susceptible to oxidative stress that can damage erythrocyte membranes,
resulting in intravascular and extravascular hemolysis.
Causes of increased oxidative stress are triggers of hemolytic crisis and include:
Antimalarial drugs (e.g., chloroquine, primaquine), sulfa
Drugs: drugs (e.g., trimethoprim-sulfamethoxazole),
nitrofurantoin, isoniazid, dapsone, NSAIDs, ciprofloxacin,
االسباب
chloramphenicol.
التي تزيد
حدة التفول Bacterial and viral Severe enzymatic deficiency can inhibit respiratory burst
وليست infections (most activity due to reduced NADPH production in phagocytes.
االسباب common cause):
.التي تسببه Inflammation: During an inflammatory reaction free radicals are
produced and can diffuse into RBCs.
Metabolic acidosis
Fava beans ()الفول.
Clinical features of G6DP deficiency:
1) Most patients are asymptomatic.
2) Recurring hemolytic crises may occur, especially following triggers:
Arise within 2–3 days after increased oxidative stress.
Sudden onset of back or abdominal pain.
Jaundice.
Dark urine.
3) Transient splenomegaly.
4) Recurrent severe infections causing symptoms of chronic granulomatous
disease (autoimmune recurrent abcess).
Confirmatory test:
Quantitative G6PD enzyme analysis.
Tests should be performed during remission (nearly about 2-3 weeks
after crisis).
Hereditary spherocytosis
Epidemiology:
Incidence: 1/5000 in the US.
Most common inherited hemolytic disease among individuals of Northern
European descent.
Etiology:
Congenital RBC membrane protein defect (mainly spectrin).
Inheritance pattern;
Autosomal dominant 75% of cases
Autosomal recessive 25% of cases
Often positive for relatives who required splenectomy
Family history and/or developed cholelithiasis (gall bladder stones) at
a young age.
Pathophysiology:
1) Genetic mutation → defects in RBC membrane proteins (especially spectrin and/or
ankyrin) responsible for tying the inner membrane skeleton with the outer lipid bilayer.
2) Continuous loss of lipid bilayer components → decreased surface area of RBCs in
relation to volume.
3) sphere-shaped RBCs with decreased membrane stability → inability to change form
while going through narrowed vessels.
Diagnostics:
MCV within normal range (80-100 fL) or slightly decreased
1) Laboratory findings;
Increased RDW, MCHC, reticulocytes, Unconjugated bilirubin and LDH.
Decreased Haptoglobin.
2) Laboratory tests
A) Eosin-5-maleimide binding test (EMA):
Test of choice.
Decreased binding between dye (eosin-5-maleimide) and RBC membrane proteins.
Binding is quantified using flow cytometry, which shows decreased mean
fluorescence.
B) Coombs test:
3) Blood smear:
Spherocytosis (small round cells without central pallor)
Treatment of Spherocytosis:
1) Non-surgical treatments:
Phototherapy and/or exchange May be necessary in neonates (e.g., to avoid
transfusions kernicterus).
Blood transfusions May be required in cases of aplastic or
hemolytic crisis.
Folic acid supplementation To maintain erythropoiesis.
Kernicterus: is a type of brain damage that can result from high levels of
bilirubin in a baby's blood.
Neonates: refers to the first 28 days after birth.
2) Splenectomy:
Sole definitive treatment ()العالج النهائي الوحيد.
Prior to splenectomy, vaccinate against Streptococcus pneumoniae,
Haemophilus influenzae type B, and Neisseria meningitides.
Complications:
Hemolytic crisis: esp. as a result of viral infection.
Aplastic crisis: following infection with parvovirus B19 (erythema infectiosum);
characterized by a low reticulocyte count (< 0.1% of total RBC count).
Megaloblastic anemia: folate and vitamin B12 deficiency may develop due to
chronic hemolysis and high RBC turnover.
Megaloblastic crisis: due to folate deficiency (although uncommon in developed
countries, it might still be seen among pregnant women).
Bilirubinate gallstone formation, possibly leading to cholecystitis, cholangitis,
and pancreatitis.
Growth retardation and skeletal abnormalities due to bone marrow expansion.
Hereditary elliptocytosis
An asymptomatic condition characterized by the
presence of elliptocytes in the blood.
Caused by mutations in genes encoding RBC
membrane proteins (e.g., spectrin, protein 4.1).
Hemoglobinopathies
An inherited mutation o the globin genes leading to a qualitative or quantitative
abnormality of globin synthesis.
a substitution of AA by another in the globin chain rate of synthesis of a globin chain is reduced:
sickle hemoglobin (HbS) Thalassemia:
hemoglobin C β-Thalassemia- reduced beta chain
hemoglobin E synthesis.
hemoglobin D α-Thalassemia- reduced alpha chain
hemoglobin O synthesis.
Hemoglobin Zurich.
1) Zeta gene is expressed only during the first few weeks of embryogensis.
Thereafter, the alpha globin genes take over.
2) The epsilon gene is expressed initially during embryogensis.
The gamma gene is expressed during fetal development.
The combination of two alpha genes and two gamma genes forms fetal
hemoglobin, or hemoglobin F.
3) Around the time of birth, the production of gamma globin declines in concert
with a rise in beta globin synthesis.
The combination of two alpha genes and two beta genes comprises the normal
adult hemoglobin, hemoglobin A.
4) The delta gene, produces a small amount of delta globin in children and adults.
The combination of two alpha genes and two delta genes forms A2 hemoglobin
Chromosome 11 Chromosome 16
Epsilon Zeta 1
Gamma Zeta 2
Delta Alpha 1
Beta Alpha 2
Hemoglobinopathies Genetic structural disorder
Due to mutation in the globin gene of hemoglobin.
Mostly autosomal recessive inheritance.
Result in hemoglobin variants with altered structure and function.
1) Altered functions include:
Reduced solubility.
Altered oxygen affinity- increased or decreased.
Methemoglobin formation (Fe 2+ Fe +3 (oxidized state) less oxygen
transport).
2) Decrease, lack of, or abnormal globin:
May be severe hemolytic anemia.
Abnormal Hb with low functionality.
Mutation may be deletion, substitution, elongation.
Epidemiology:
Predominantly affects individuals of African and East Mediterranean descent.
HbS gene is carried by 8% of the African American population.
Sickle cell anemia is the most common form of intrinsic hemolytic anemia
worldwide.
Pathophysiology:
Pathomechanism:
HbS polymerizes when deoxygenated, causing deformation of erythrocytes
(“sickling”).
This can be triggered by any event associated with reduced oxygen
tension.
GLU is polar AA, and thus hydrophilic, whereas the Val is
hydrophobic…leading to stickness of the hemoglobin in the deoxyform.
Hypoxia (e.g., at high altitudes).
In homozygotes, up to 100% of the hemoglobin molecules are affected, leading to
sickle cell formation under minimally decreased oxygen tension.
Infections.
Dehydration.
Sudden changes in temperature.
Stress.
Sickle cells lack elasticity and adhere to vascular endothelium, which disrupts
microcirculation and causes vascular occlusion and subsequent tissue
infarction.
Extravascular hemolysis and intravascular hemolysis are common and result
in anemia.
The body increases the production of fetal hemoglobin (HbF) to compensate
for low levels of HbA in sickle cell disease.
2) Aplastic crisis;
Severe drop in hemoglobin and associated reticulocytopenia due to an
infection with parvovirus B19.
Can temporarily suppress bone marrow erythropoiesis.
C) Infection:
Pneumonia.
Osteomyelitis; most common cause: Salmonella.
Sepsis; most common cause: Streptococcus pneumoniae.
D) Vaso-occlusive events:
Vaso-occlusive crises (painful episodes, painful crisis): recurrent episodes of
severe deep bone pain and dactylitis { }التهاب نهايات األصابع most common
symptom in children and adolescents ()المراهقين.
Acute chest syndrome.
Priapism ()الم في القضيب نتيجة االنتصاب لفترة طويلة.
Stroke.
Infarctions of virtually any organ (particularly spleen) and avascular necrosis
with corresponding symptoms.
Chronic symptoms:
Chronic hemolytic anemia: fatigue, weakness, pallor: usually well-tolerated.
Chronic pain.
Cholelithiasis (pigmented stones).
Symptoms of other forms of sickle cell syndrome (HbSC disease and HbS/beta-
thalassemia) are similar to sickle cell anemia but less severe.
Sickle cell disease with drepanocytes (sickle cell) and target cells
morphologies in sickle cell disease.
Complications:
1) Organ damage:
Recurrent vascular occlusion and disseminated infarctions lead to progressive
organ damage and loss of function.
homozygotes High morbidity and mortality ()ارتفاع االغتالل والوفيات.
heterozygotes Organ damage is very rare.
2) Functional asplenia.
3) Renal papillary necrosis.
4) Avascular osteonecrosis.
5) Recurrent strokes.
6) Priapism.
7) Chronic lung disease, acute chest syndrome.
8) Cardiomyopathy (cardiomegaly) Heart failure.
9) Acute sickle hepatic crisis.
10) Hand-foot syndrome (in small, i.e., around age of 3y);
Treatment {Long term management}:
1) Prevent infections:
Pneumococcal vaccines.
Meningococcal vaccines.
Daily penicillin prophylaxis; at least until the age of 5 years.
If sepsis is suspected, treat with IV third-generation cephalosporin (e.g., ceftriaxone).
Curative therapy:
Allogeneic bone marrow transplantation:
Used in; homozygotes, children <16 years with severe disease.
Hemoglobin C Disease:
Is an inherited (autosomal recessive) blood disorder that that may cause a person
to have mild anemia (low blood count).
Is a common hemoglobin variant that has a single amino acid substitution (lysine
substituted for the glutamate) in the sixth position of the beta-globin chain.
hemoglobin C trait (HbAC) Hemoglobin C disease (HbCC)
phenotypically normal May have chronic hemolytic anemia.
Hemoglobin E (HbE)
An abnormal hemoglobin with a single point mutation in the β chain. At
position 26 there is a change in the amino acid, from glutamic acid to lysine
(E26K).
E26K; E = Glutamate, 26 = NO.AA. K = Lysine
Is a common but minor blood abnormality.
The blood disorder is often identified because there are slight abnormalities in
the size and appearance of the red blood cells.
2) Bleeding disorders:
A group of disorders characterized by defects in hemostasis, which leads to an
increased susceptibility to bleeding.
Caused either by:
Platelet disorders (primary hemostasis defect).
Coagulation defects (secondary hemostasis defect):
Combination of both.
Coagulation defects may be further divided into intrinsic or extrinsic defects
according to the pathway of the coagulation cascade affected.
Etiology
2) Extrinsic pathway:
Factor VII deficiency.
3) Both pathways:
A) Deficiency or inhibition of vitamin K-dependent coagulation factors (II, VII, IX, and X)
--- {1972}.
Clinical feature:
1) Blood in urine or stool.
2) Frequent, large bruises.
3) Heavy bleeding after giving birth.
4) Excessive bleeding: that does not stop with pressure and may start spontaneously,
such as with nosebleeds, or bleeding after a cut, dental procedure, or surgery.
5) Heavy menstrual bleeding: which includes menstrual bleeding that often lasts longer
than seven days or requires changing sanitary pads or tampons more than every hour.
6) Petechiae: bleeding under the skin causing tiny purple, red, or brown spots.
7) Redness, swelling, stiffness, or pain from bleeding into muscles or joints, which is
particularly common with inherited hemophilia.
8) Umbilical stump bleeding: that lasts longer than what is typical for newborns— about
one to two weeks after the umbilical cord is cut— or that does not stop.
Complications:
Severe bleeding disorders can cause serious and life-threatening problems,
including:
Bleeding in the CNS hemorrhagic stroke
Bleeding in the throat Swelling and block the windpipe.
Bleeding into the abdomen Inflammation and damage to nerves.
From bleeding into joints overtime, especially
Damaged joints for people who have inherited hemophilia
This can also cause chronic pain.
Hard masses in the bones From pooled blood.
Miscarriages اإلجهاض
If the number of platelets is too low, you may have a platelet disorder instead of a
clotting factor disorder.
4) Mixing test:
Measure the amount of von Willebrand factor, whether the factors are working
correctly, or which type of VWD you have.
Determine whether certain clotting factors are missing or show up at lower levels
than normal, which can indicate the type and severity of the bleeding disorder.
For example, if you have very low levels of clotting factor VIII, you may
have hemophilia A.
7) Bethesda test:
9) Genetic testing:
Clinical features
Type 1 and avWD usually manifest more mildly; type 3 is the most severe form.
1) Often asymptomatic.
2) Symptomatic individuals may develop the following symptoms:
A) Bleeding after surgical procedures or tooth extraction
B) GI bleeding (can be caused by angiodysplasia)
C) Menorrhagia (affects up to 92% of women with vWD) [5]
D) Severe cases: large hematomas, hemarthrosis, life-threatening bleeding
(e.g., during childbirth).
E) Mucocutaneous bleeding:
Ecchymoses, easy bruising
Epistaxis
Bleeding of gingiva and gums
Petechiae
Prolonged bleeding from minor injuries
Diagnostics
1) History:
Recurrent episodes of bleeding since childhood
Often positive family history.
2) Laboratory studies:
High bleeding time.
High or normal aPTT.
Ristocetin: drug aid
Normal PT and platelet count.
platelet aggregation.
Decrease in vWF and VIII factor.
Ristocetin cofactor level < 30 IU/dL.
Treatment of VWFDs:
Hemophilia
Disorders of blood clotting and consequently may lead to serious bleeding. In
the majority of cases, these disorders are hereditary.
Hemophilia is caused by an X-linked recessive defect (inherited or
spontaneous mutation) or antibody production against clotting factors.
Etiology:
Type Deficiency Percent
Hemophilia A Factor VIII 80 %
Hemophilia B Factor IX 20 %
very rare (increased frequency in the Ashkenazi
Hemophilia C Factor XI Jewish population); caused by an autosomal
recessive defect
1) Spontaneous bleeding or delayed-onset bleeding (joints, muscular and soft tissue, and
mucosa) in response to different degrees of trauma:
Hemarthrosis
Diagnostics:
1) Patient and family history.
2) Screening.
PT aPTT Platelet count
Normal Usually prolonged Normal
Treatment
A) Substitution of clotting factors:
Etiology:
Infections Sepsis (esp. G –Ve)
Trauma Acute traumatic coagulopathy.
Burns.
Obstetric Amniotic fluid embolism.
complications Abruptio placenta
Organ failure Acute pancreatitis.
Acute respiratory distress syndrome (ARDS).
Acute promyelocytic leukemia (APL), acute myelocytic leukemia.
Malignancies Solid tumors, e.g.:
Pancreatic, Ovarian, Gastric and Non-small cell lung cancer.
Toxins Snake bites
Acute hemolytic transfusion reaction (AHTR).
Immunologic Transplant reaction (e.g., graft-versus-host disease).
Extracorporeal procedures (e.g., dialysis).
Vascular Aortic aneurysms.
malformations
Dilution Massive transfusion
Nephrotic syndrome.
Other New thrombus formation.
Hemolysis.
Acidosis.
Drug reactions -----------------------------------------------------------------------------------------
Pathophysiology:
1) Underlying disease (↑) tissue factor (III) (TF) presentation (e.g., due to increased
expression after trauma) (↑) activation of thrombin generation of fibrin
consumption of natural anticoagulants (e.g., antithrombin, thrombin-antithrombin
complex (TAT), protein C).
Clinical features:
1) Bleeding:
Hematemesis, hematochezia.
Hematuria.
Oozing of blood from surgical wounds or intravenous lines.
Petechiae, purpura, ecchymoses.
Treatment
1) Treatment of the underlying disease is the core of management.
2) Blood products:
Prophylactic heparin.
Therapeutic heparin.
Other coagulation inhibitors: Consider in individual cases in consultation with
a specialist.
Cryoprecipitate
A product obtained from frozen blood
plasma via centrifuge.
Inherited platelet disorders
Platelet membrane glycoproteins and their function: An overview:
Platelet membrane glycoproteins are surface glycoproteins found on platelets
(thrombocytes) which play a key role in hemostasis. When the blood vessel wall
is damaged, platelet membrane glycoproteins interact with the extracellular
matrix.
Membrane glycoproteins:
Glycoprotein Ib-IX-V complex (GPIb-IX-V):
The deficiency in glycoprotein Ib-IX-V complex synthesis leads to Bernard-
Soulier syndrome.
Glycoprotein VI (GPVI):
Is an important collagen receptor involved in collagen-induced platelet activation
and adhesion.
It plays a key role in then procoagulant activity and subsequent thrombin
and fibrin formation.
Pathophysiology:
There are specific GP receptors on the platelet membrane, which function in platelet
adhesion, activation, and aggregation.
The GP (Ib-IX-V) receptor complex is responsible for platelet adhesion through its
interaction with von Willebrand factor on the exposed subendotlielium.
The GP (Ib-IX-V) receptor complex is composed of four transmembrane polypeptide
subunits-disulfide-linked alpha and beta subunits of GPIb, and noncovalently bound
subunits GPIX and GPV.
The platelets of BSS cases lack or have a dysfunctional GPIb-IX-V receptor.
This results in defective adhesion to the subendotlielium.
Clinical manifestations:
Manifest with a tendency to bleed in early childhood.
Mucocutaneous bleeding is seen predominantly.
Easy bruising, purpura, epistaxis, bleeding gums, menorrhagia, and excessive bleeding
after surgery or trauma are common symptoms.
Diagnosis:
Although thrombocytopenia is generally observed in BSS, the number of platelets is
variable.
Giant platelets are seen in peripheral blood smear.
Bleeding time found to be prolonged. Routine coagulation tests should be found normal.
In vitro platelet aggregation studies characteristically indicate that aggregation with
ristocetin failed and responded slowly with low doses of thrombin.
Flow cytometric analysis of platelet: defective binding with CD42a (GPIX), CD42b (GP
lb), CD42c (GP lb), and CD42d (GPV).
Treatment:
1) BSS treatment is generally supportive.
2) Platelet transfusion is used to treat when
surgery is needed or when there is a risk of
life-threatening bleeding.
The patient may develop antiplatelet
antibodies due to the presence of
glycoproteins Ib/IX/V, which are
present
on the transfused platelets but absent
from the patient’s own platelets.
Glanzmann thrombasthenia
GT is an autosomal recessive congenital bleeding disorder characterized by a lack of
platelet aggregation due to defect and/or deficiency of (𝜶Ilb𝜷3) integrin.
Patients with this disorder often experience lifelong bleeding episodes involving
mucocutaneous membranes.
Etiology:
GT is usually caused by decreased or absent expression of 𝜶Ilb or 𝜷3, abnormalities in
protein folding, transport of the integrin subunit causing post-translational defective
processing or decreased surface expression, or abnormalities affecting protein function.
Pathophysiology:
Diagnosis:
The diagnosis of GT is often not noticed, because many platelet disorders share common
clinical and laboratory features.
Treatment:
A gradual treatment standard is applied in GT treatment. The first treatment for mild
bleeding is local measures including local compression, cauterization, stitching, or ice
therapy.
The treatment applied in case of unresponsiveness to these treatments or in heavier
bleeding is antifibrinolytic therapy first, followed by platelet transfusion, and recombinant
active factor VII (rFVIIa) if bleeding persists.
Immune thrombocytopenia (Immune thrombocytopenic purpura)
Primary immune thrombocytopenia Secondary immune thrombocytopenia
An autoimmune disorder characterized An autoimmune hematologic disorder
by isolated thrombocytopenia causing isolated thrombocytopenia
(< 𝟏𝟎𝟎, 𝟎𝟎𝟎/𝒎𝒎𝟑) with no known that is secondary to an identifiable
precipitating cause. trigger.
Etiology:
Primary ITP: idiopathic (most common).
Pathophysiology:
Antiplatelet antibodies (mostly IgG directed against, e.g., GpIIb/IIIa, GpIb/IX) bind
to surface proteins on platelets sequestration by spleen and liver (↓) platelet
count bone marrow megakaryocytes and platelet production increase
in response (in most cases).
Clinical features:
Most commonly:
A Symptomatic.
Splenomegaly is typically absent.
Diagnostics:
ITP is a diagnosis of exclusion; patients typically have a low platelet count with no other
abnormalities.
Etiology:
1) ADAMTS13 deficiency/inhibition:
Diagnostics:
Hematology
(↓) Platelets
(↓) Hemoglobin
(↑) Reticulocytes
(↓) Haptoglobin
Normal or mildly prolonged prothrombin time (PT) and activated partial
thromboplastin time (aPTT)
Negative Coombs test.
Serum chemistry
Identification of secondary causes (e.g., tests for pregnancy, SLE, HIV, malignancy)
Treatment:
1) Monitoring and correction;
Electrolyte disturbances.
RBC transfusions.
2) Prompt initiation of plasma exchange therapy (PEX).
3) Glucocorticoids.
4) Rituximab is reserved for severe cases.
Neonatal thrombocytopenia
Neonatal thrombocytopenia is defined as a platelet count <150,000/ 𝝁L.
Although severe neonatal thrombocytopenia (defined as a platelet count <50,000/𝝁L) is
uncommon in the general healthy newborn population, the risk increases for infants
admitted to the neonatal intensive care unit (NICU) especially for the extremely preterm
infants (birth weight [BW] <1000 g or gestational age [GA] < 28 weeks).
Presentation
The clinical presentation of neonatal thrombocytopenia includes patients detected
incidentally ( )بالصدفةby a low platelet count from a complete blood count (CBC) obtained
for other reasons, at-risk patients identified by a screening CBC, and symptomatic
infants with evidence of bleeding (eg. petechiae, large ecchymoses, cephalohematoma,
or oozing from the umbilical cord or puncture sites).
Diagnostic evaluation
The diagnostic evaluation is focused on determining and, if possible, directing specific
therapy to the underlying cause of neonatal thrombocytopenia.
However, establishing a diagnosis can be challenging because of the overlap of the
clinical presentation among different conditions, and because frequently there may be
multiple potential causes.
The diagnostic approach to neonatal thrombocytopenia is centered on the timing of
presentation (early within the first 72 hours of life or later), the severity of
thrombocytopenia, the infant's clinical condition, and the maternal and neonatal history,
including labor and delivery.
Pathophysiology:
The major mechanism is impaired platelet production.
Management approach:
A) In the majority of cases, neonatal thrombocytopenia resolves without intervention.
B) Specific therapy, if available, should be given to patients in whom an etiology has
been identified (e.g., sepsis).
C) Platelet transfusion:
Indications; Most platelet transfusions in neonates are given prophylactically to patients
without evidence of bleeding.
Indications for Platelet Transfusion in Neonates:
In extracorporeal membrane oxygenation (ECMO), blood is pumped outside of your body to a heart-
lung machine that removes carbon dioxide and sends oxygen-filled blood back to tissues in the body
Mechanism:
PTP is rare, but usually occurs in women who have had multiple pregnancies
or in people who have undergone previous transfusions.
The precise mechanism leading to PTP is unknown.
The most commonly occurs in individuals whose platelets lack the HFA-1a
antigen (old name: PLA1).
The patient develops antibodies to the HPA-1a antigen leading to platelet
destruction.
Treatment:
1) Symptoms are usually sudden in onset and self-limiting, most often resolving
within 2 weeks.
2) IVIG therapy is the primary treatment.
3) Additionally, PTP is an American Society for Apheresis Category III indication
for plasmapheresis.
Hypercoagulable disorders
The main causes of thrombosis:
1. Turbulence flow (Stasis).
2. Endothelial injury.
3. Hypercogulation.
Endothelium, the inner most single layer of cells lining the blood vessels,.
Endothelium functions:
1) Surface for thrombosis formation and critically regulates blood fluidity and
homeostasis.
2) Barrier which separates blood clotting factors from exposure to subendothelial
prothrombotic extracellular matrix components.
3) Secretes or expresses vasoactive factors that modulate platelet reactivity,
coagulation, fibrinolysis, and vascular contractility, all of which contribute to
thrombotic formation.
Such factors include nitric oxide, prostacyclin, Von Willebrand factor
(VWF), thrombomodulin, endothelin, etc.
Antithrombin III, protein C, and protein S. naturally occurring
anticoagulant proteins
protein C and protein S Antithrombin III
Inhibiting the action of the cofactors factor inhibits the serine proteases (factors II, IX,
Va and factor VIlla. X, XI, and XII)
Clinical features:
Venous thromboembolism Arterial thromboembolism
Deep venous thrombosis (DVT),
thromboembolism pulmonary embolism (PE), portal Ischemia.
vein thrombosis (PTV), cerebral Acute coronary syndrome.
venous thrombosis (CVT).
Onset at age <50 years of
either of the following:
Unprovoked VTE.
VTE associated with only
thrombophilia weak risk factors. In a young patient with no
Unusual thrombus localization. cardiovascular risk factors.
Prevalence
Heterozygosity: Homozygosity:
∼ 5% < 1%
Normal Pathological
Activated protein C (APC) In such patients, Gln506-Va is resistant
inactivates factor V in the clotting to cleavage by APC → factor V remains
cascade → decreases the activation active → activates prothrombin →
of thrombin. increases thrombotic events (e.g.,
peripheral and cerebral vein thrombosis,
A DNA point mutation substitutes recurrent pregnancy loss).
guanine for adenine → corresponding
mRNA codon forms glutamine in Risk of thromboembolism is several
place of arginine on position 506 times higher in patients with homozygous
(Arg506Gln mutation) near the mutations than in those with heterozygous
polypeptide cleavage site of factor V. mutations.
Pathophysiology Prevalence
In combination with factor IXa, factor VIIIa activates factor X → ∼ 5%
increases thrombotic events.
3) Prothrombin mutation:
6) Hyperhomocysteinemia:
Pathophysiology of acquired thrombophilia
Etiology Pathophysiology
Extended immobilization during procedure → blood stasis.
Surgery
Vessel instrumentation → endothelial damage.
Results in decreased venous blood flow, immobilization (blood
Trauma stasis), and release of tissue factor (hypercoagulability) → increased
clotting.
Cancers excrete procoagulant factors (e.g., tissue factor and cancer
Malignancy procoagulant).
The risk of thromboembolism is highest during first hospitalization
and initiation of chemotherapy.
Immobilization Prolonged immobilization (e.g., extended travel, hospitalization, bed
rest) → increased venous stasis.
Causes endothelial damage.
Smoking The risk is significantly higher in women who also use oral
contraceptives.
Leads to chronic systemic inflammation and impaired fibrinolysis.
Obesity The risk of thromboembolism increases with increasing Body Mass
Index (BMI).
Acquired antibodies directed against plasma proteins bound to
phospholipids (e.g., lupus anticoagulant, anti-cardiolipin, beta2-
Antiphospholipid glycoprotein I antibodies) → aggregation of plasma proteins (e.g.,
syndrome clotting factors) → induces venous and arterial clotting →
miscarriages, DVTs, portal vein thrombosis, and strokes.
Associated with SLE and rheumatoid arthritis.
Nephrotic syndrome Loss of plasma antithrombin in urine and an increase in blood
viscosity due to extravasation of fluid from albumin loss in urine.
Oral contraceptive
pills (OCPs) or Increased estrogen and progestin → increase in prothrombin and
hormone fibrinogen and a decrease in protein S.
replacement therapy
(HRT)
Heparin-induced Antibodies against platelet factor 4 (PF-4) → increased activation of
thrombophilia platelets (hypercoagulability) and a depletion of platelets
Clotting factors increase (hypercoagulability).
Pregnancy Protein C and protein S decrease.
Venous stasis as the uterus enlarges.
Progressive endothelial damage.
Increase in pro-clotting factors without a concomitant increase in
Advanced age protein C.
Increase in other pro-clotting comorbidities (e.g., malignancy).
Decreased physical activity.
2) Thrombophilia testing
Hereditary thrombophilia Acquired thrombophilia
Activated protein C resistance assay. Antiphospholipid antibody panel:
Prothrombin G20210A mutation testing. Lupus anticoagulants.
Activity assays for protein C, protein S, and anticardiolipin antibodies).
antithrombin.
Treatment
1) Standard management of thromboembolic diseases:
Prevention:
Standard VTE prophylaxis is indicated in select circumstances regardless of
thrombophilia status (e.g., postoperative status, prolonged immobilization or
hospitalization, active malignancy).
Acute leukemia
Acute leukemia are malignant neoplastic diseases that arise from either the
lymphoid or myeloid cell line.
In acute leukemia, the peripheral blood has decreased mature forms and
increased immature forms “blasts”, which have immature chromatin with
nucleoli.
The bone marrow has increased immature cells (blasts).
Acute symptoms are secondary to marrow failure, which can produce
decreased erythrocytes (causing anemia and fatigue), decreased leukocytes
(permitting infections and fever), and decreased platelets (inducing bleeding).
There are two main types of acute leukemia:
Acute lymphoblastic leukemia (ALL) acute myeloid leukemia (AML)
most common childhood malignancy Primarily affects adults.
Down syndrome: The risk of AML is, like that of ALL, 10–20 times higher in
patients with Down syndrome compared to the general population.
Classification of ALL:
French-American-British (FAB).
Based on morphologic and genetic factors.
Based on the origin (B cell or T cell) and maturity of the leukemic cells.
Classification of AML:
The French-American-British (FAB) - according to the histopathological
appearance of the cells:
For myoblast in general M0-AML Acute myeloblastic leukemia without maturation
M1-AML Acute myeloblastic leukemia with minimal granulocyte
For granulocytes maturation.
M2-AML Acute myeloblastic leukemia with granulocyte maturation.
M3-AML Acute promyelocytic leukemia (APL).
For monocytes M4-AML Acute myelomonocytic leukemia.
M5-AML Acute monocytic leukemia.
For erythrocytes M6-AML Acute erythroid leukemia.
For thrombocytes M7-AML Acute megakaryoblastic leukemia.
Pathophysiology
Clinical features:
Clinical features are either related to bone marrow failure, infiltration of organs by
leukemic cells, or a combination of both.
Sudden onset of symptoms and rapid progression (days to weeks).
Anemia: fatigue, pallor, weakness.
Thrombocytopenia: epistaxis, bleeding gums, petechiae, purpura.
Immature leukocytes: frequent infections, fever.
Hepatosplenomegaly (caused by leukemic infiltration).
Oncologic emergencies can be the first sign of leukemia, e.g., an elderly
patient presenting with priapism or DIC may have leukostasis (more common
in AML than ALL): See oncologic emergencies for further details.
Clinical features of ALL:
1) Painless lymphadenopathy.
2) Bone pain (presenting as limping or refusal to bear weight in children).
3) Airway obstruction (stridor, difficulty breathing) due to mediastinal or thymic
infiltration (primarily in T-cell ALL).
4) Features of SVC syndrome (SVC compression).
5) Meningeal leukemia (or leukemic meningitis); headache, neck stiffness, visual
field changes, or other CNS symptoms (caused by CNS involvement).
6) Testicular enlargement (rare finding).
7) B symptoms: Fever, night sweats, unexplained weight loss.
Pathophysiology:
1) Tumor cell lysis intracellular component (K+, PO4-, nucleic acid) released
into blood (↑) nucleic acid conversion to uric acid hyperuricemias
urate nephropathy and risk of acute kidney injury.
2) (PO4-) binds with calcium decrease calcium (ca+2) hypocalcemia
neuronal excitability risk of seizures.
3) Hyperkalemia hyperpolarization cardiac arrhythmias.
Cardiac arrhythmias.
Seizures, epilepsy.
Kidney injuries.
Hypocalcemia.
Hyperkalemia, hyperuricemia and (↑)𝑷𝑶𝟒−𝟐.
2) Leukostasis:
A medical emergency characterized by tissue hypoxia and hypercoagulability
due to an excessive number of immature leukocytes causing microvascular
obstruction.
Diagnosis of leukemia:
Cell morphology
ALL AML
Size Small- to intermediate-sized blasts. Large blasts (2–4 times the size of an RBC).
Nucleus Blasts with large, irregular nuclei Blasts with round or kidney-shaped nuclei that
(high nuclear-cytoplasmic ratio). contain more cytoplasm.
Clarity Inconspicuous nucleoli. Prominent nucleoli.
Granules Coarse granules. Fine granules.
Some subtypes (especially M3, or APL)
exhibit Auer rods:
Pink-red, rod-shaped granular inclusion
Auer rods No Auer rods. bodies in malignant immature myeloblasts
or promyelocytes..
Myeloperoxidase (MPO) positive.
Leukemic hiatus: the presence of blasts and
Others ------------------------------------------------ mature leukocytes but no intermediate forms.
3) Specialized studies:
A) Immunophenotype:
Immunohistochemistry
ALL AML
MPO Negative. positive
Terminal deoxynucleotidyl transferase (TdT) Positive. negative
Periodic acid-Schiff (PAS): often positive negative
Most common in
chronic leukemia
B) Genetic studies
Prognosis:
5-year survival rate following treatment; Five-year life expectancy for a patient
with leukemia.
ALL generally higher compared to AML.
AML 30% (80% for elderly people and 20% for children).
Lymphoma
Is a cancer of the lymphatic system, which is part of the body's germ-fighting
network.
The lymphatic system includes the lymph nodes (lymph glands), spleen,
Thymus gland and bone marrow.
Lymphoma can affect all those areas as well as other organs throughout
the body.
The main subtypes of lymphoma:
Hodgkin's lymphoma (formerly called Hodgkin's disease).
Non-Hodgkin's lymphoma.
Hodgkin lymphoma (Lymphogranulomatosis)
Hodgkin lymphoma (HL) is a malignant lymphoma that is typically of B-cell
origin.
The incidence of HL has a bimodal age distribution, with peaks in the 3rd and
6th–8th decades of life.
Etiology
The exact causes are unknown, but several risk factors have been associated
with HL:
Strong association with Epstein-Barr virus (EBV).
Immunodeficiency: e.g., organ or cell transplantation, immunosuppressant,
HIV infection , chemotherapy.
Autoimmune diseases (e.g., rheumatoid arthritis, sarcoidosis).
Clinical features
1) Painless lymphadenopathy
4) B symptoms
Night sweats, weight loss > 10% in the past 6 months, fever > 38°C (100.4°F)
Can occur in a variety of diseases In the case of confirmed HL, the presence of a
single B symptom suffices for a positive diagnosis of B symptoms.
5) Pel-Ebstein fever:
Intermittent fever with periods of high temperature for 1–2 weeks, followed by a
febrile periods for 1–2 weeks.
Relatively rare but very specific for HL.
6) Alcohol-induced pain:
Diagnostics
Diagnosis of HL is primarily based on medical history and clinical features (B
symptoms, localization of lymph node involvement) and is confirmed with lymph
node biopsy.
Nodules of
Reed-Sternberg Nodules with Presence of Numerous
cells within numerous Reed- Reed-Sternberg Reed-Sternberg
lacunae, Sternberg cells, cells. cells.
Pathology separated by
collagenous Histiocytes, Reactive Decreased
tissue with eosinophils, and lymphocytosis number of
sclerosing plasma cells. that causes lymphocytes.
appearance. distortion of the
Lymphocyte lymph node
rich. architecture.
Prognosis
Good prognosis:
Etiology
1) Various viruses have been attributed to different types of NHL.
Epstein-Barr virus, a endemic variant of Burkitt lymphoma
DNA virus
Human T-cell leukemia adult T-cell lymphoma
virus type 1 (HTLV-1)
Hepatitis C virus (HCV) Clonal B-cell expansions.
Helicobacter Increased risk of gastric mucosa-associated lymphoid
pylori infection tissue (MALT) lymphomas, a primary gastrointestinal
lymphoma.
2) Drugs like;
Phenytoin, digoxin, TNF antagonist are also associated with Non-Hodgkin
lymphoma.
3) Environmental factors:
Aromatic hydrocarbons (e.g., benzene, associated for AML), radiation.
4) Congenital immunodeficiency: increased risk of
Wiskott-Aldrich syndrome.
Severe combined immunodeficiency disease (SCID).
Patients with AIDS (Acquired immunodeficiency syndrome) can have
primary CNS lymphoma.
5) The autoimmune disorders:
Like Sjögren syndrome, rheumatoid arthritis, and Hashimoto thyroiditis are
associated with an increased risk of NHL.
B-cell lymphomas (85% of all NHLs):
Indolent (low-grade) Aggressive (high-grade)
Hairy cell leukemia. Burkitt lymphoma.
Follicular lymphoma. Precursor B-cell lymphoblastic
Marginal zone B-cell lymphomas (MZLs). lymphoma.
Waldenstrom macroglobulinemia. Diffuse large B-cell lymphoma.
Small lymphocytic lymphoma (SLL). Mantle cell lymphoma.
Indolent (low-grade)
Waldenstrom macroglobulinemia:
IgM antibodies
Aggressive (high-grade)
Burkitt lymphoma
Most common in children.
Translocation t(8;14) in 75% of cases.
Starry sky pattern
Microscopic finding that resembles a starry sky
2) Sezary syndrome:
Extranodal disease:
Excisional tissue biopsies are recommended.
Histopathology and specialized studies
These studies help determine the subtype of NHL.
Histopathology:
Provides a detailed morphology of individual proliferating cells and a description of the
pattern of lymph node (or tissue) infiltration (e.g., nodular, diffuse).
Immunophenotype (e.g., flow cytometry, immunohistochemistry)
Detects surface antigens,
Determines the specific cell type (B cell/T cell), and identifies specific markers Possible
findings include:
B-cell lymphomas: CD20 positive
T-cell lymphomas: CD3 positive
Genetic studies
• Cytogenetics (karyotype, FISH): can identify chromosomal abnormalities.
Treatment
Most patients will receive treatment with systemic chemotherapy and/or radiotherapy.
Selection of treatment: based on the subtype of NHL, staging, and prognosis;
Risk factors
Advanced age
Environmental factors: organic solvents
Family history
Pathophysiology
Acquired mutations in hematopoietic stem cells → increased proliferation of
leukemic B cells with impaired maturation and differentiation in the bone marrow,
resulting in:
Suppression of the proliferation of normal blood cells
Immunosuppression.
Hypogammaglobulinemia.
Granulocytopenia.
Thrombocytopenia.
Anemia.
Infiltration of the lymph nodes, liver, and spleen.
Clinical features
About half of cases of CLL remain asymptomatic for a long period, resulting in
late or incidental diagnosis.
1) Weight loss, fever, night sweats, fatigue (B symptoms)
2) Painless lymphadenopathy
3) Hepatomegaly and/or splenomegaly may occur.
4) Repeated infections
Severe bacterial infections (e.g., necrotic erysipelas)
Mycosis (candidiasis)
Viral infections (herpes zoster)
Symptoms of anemia and thrombocytopenia
5) Dermatologic symptoms
Leukemia cutis.
Chronic pruritus.
Chronic urticaria.
Complications
1) Immunosuppression with subsequent infections (most common cause of
death)
2) Secondary malignancies
3) Hyperviscosity syndrome
3) Autoimmune hemolytic anemia (of both the warm and cold agglutinin type)
4) Richter transformation or Richter syndrome: transformation into a high-grade
NHL (usually diffuse large B cell lymphoma)
Occurrence: ∼ 5% of cases
Diagnostic indicators:
Rapidly progressive lymphadenopathy → lymph node biopsy required.
New onset of B symptoms.
↑ LDH.
Diagnostics
1) CBC:
Persistent lymphocytosis with a high percentage of small mature lymphocytes
Findings that indicate suppression of normal myelopoiesis:
Granulocytopenia
Low RBC count (due to autoimmune hemolysis)
Low platelet count
3) Flow cytometry:
Detection of B-CLL immunophenotype (CD5, CD19, CD20, CD23), light chain
restriction (kappa or lambda)
5) Blood smear:
Smudge cells (Gumprecht shadows) – mature lymphocytes that rupture easily
and appear as artifacts on a blood smear
Principles of treatment
1) Asymptomatic CLL: observe and monitor disease progression
2) Symptomatic CLL or advanced stage (Rai stage > 0, accelerated disease
progression)
• Chemotherapy
• If CD 20 positive: rituximab
• Targeted therapy with ibrutinib
3) Refractory CLL or early recurrence in fit, young patients:
Allogeneic stem cell transplantation
Prognostic factors
• Advanced age is associated with a poor overall survival rate.
2) Peripheral blood
The neoplastic lymphocytes seen in this disease are large in size with azurophilic
granules that contains proteins involved in cell lysis such as perforin and granzyme B
3) Bone marrow
The lymphocytic infiltrate is usually interstitial, but a nodular pattern rarely occurs.
4) Immunophenotype
The neoplastic cells of this disease display a mature T-cell immunophenotype, with the
majority of cases showing a;
CD4 CD8
- Ve + Ve
Genetic findings
Clonal rearrangements of the T-cell receptor (TCR) genes are a necessary condition for
the diagnosis of this disease.
Mycosis fungoides
Mycosis fungoides is an indolent, CD4+ cutaneous T-cell lymphoma that presents on
the skin.
It is characterized by scaly, pruritic, well-demarcated skin plaques and patches.
Clinical features
Initially, pruritic cutaneous plaques, patches, and brownish nodules develop.
Subsequently, systemic spread occurs, including lymphadenopathy and
hepatosplenomegaly.
Sézary syndrome
A cutaneous T-cell lymphoma with leukemic dissemination of mutated T cells
Epidemiology
Exact prevalence unknown
Can be an advanced form of mycosis fungoides or
arise de novo
Clinical features
Systemic skin lesions
Erythroderma accompanied by palmar and plantar
hyperkeratosis
Intense pruritus
Generalized lymphadenopathy
Diagnostics:
Based on the characteristic triad of pruritic erythroderma,
lymphadenopathy, and atypical T cells (Sézary cells) on peripheral blood smear.
Clinical features:
Generalized lymphadenopathy
Hepatosplenomegaly
Skin lesions: may be similar to those seen in mycosis fungoides
Lytic bone lesions.
Diagnostics
• Peripheral blood smear: lymphocytes with condensed chromatin and
hyperlobulated nuclei that resemble clover leaves.
• Laboratory tests: hypercalcemia, (↑) LDH
• Immunophenotype: stain positive for CD2, CD4, CD5, CD29, and CD45RO
Plasma cell tumors (Multiple myeloma)
Plasma cell dyscrasia:
Plasmacytoma:
Multiple myeloma:
Pathophysiology
Neoplastic proliferation of plasma cells
Bone marrow infiltration by malignant plasma cells → suppression of
hematopoiesis → leukopenia, thrombocytopenia, and anaemia.
Cell proliferation → osteolysis → hypercalcemia.
Overproduction of monoclonal immunoglobulin and/or light chains →
dysproteinemia (a state of pathologically increased synthesis of
immunoglobulins and/or their subunits) → kidney damage (e.g., myeloma cast
nephropathy) and/or paraprotein tissue deposition (may cause amyloidosis)
Nonfunctioning antibodies → functional antibody deficiency
↑ Serum viscosity → hyperviscosity syndrome.
Clinical features
Often asymptomatic
Mild fever, night sweats, weakness, and weight loss (B symptoms).
back bone pain (most common symptom)
Symptoms of hypercalcemia
Spontaneous fractures
Foamy urine (caused by Bence Jones proteinuria).
Renal insufficiency Increase risk of amyloidosis.
Suppression of hematopoiesis → leukopenia, thrombocytopenia, and anaemia.
Increased risk of infection
Increased risk of petechial bleeding.
Diagnostics
The following tests are required for patients with suspected multiple myeloma
(MM):
Serum protein electrophoresis or free light chain assay (best initial test)
24-hour urine protein electrophoresis
Bone marrow biopsy (confirmatory test)
Laboratory tests (CBC and biochemistry) to assess for hypercalcemia, anemia
and renal insufficiency
Imaging to assess for bone lesions.
Laboratory tests
CBC
rouleaux formation
Biochemistry
Urinalysis
Systemic manifestations
AL amyloidosis:
Infections
Immunodeficiency (nonfunctional immunoglobulins) and side effects of
medications
Diagnostics
Usually, an incidental finding on workup (e.g., protein electrophoresis) for
other conditions such as vasculitis, hypercalcemia, skin rashes, peripheral
neuropathy, increased ESR, and hemolytic anemia
Risk of progression to myeloma
Determined by the following predictors:
Presence of non-immunoglobulin G-type M protein.
M-protein concentration ≥ 1.5 g/dL.
abnormal serum free light chain (SFLC) ratio
M protein
1. A monoclonal immunoglobulin (paraprotein) that is produced in plasma cell
dyscrasias (e.g., multiple myeloma, Waldenstrom macroglobulinemia,
monoclonal gammopathy of undetermined significance).
Detected on serum or urine electrophoresis as a spike in the gamma-
globulin zone (monoclonal spike, M spike).
2. A virulence factor of group A streptococci that prevents opsonization by
complement factor C3.
Waldenstrom macroglobulinemia
A type of non-Hodgkin lymphoma associated with abnormal production of
monoclonal IgM antibodies.
Clinical features:
Peripheral neuropathy
Impaired platelet function
Formation of cold agglutinins (IgM) with hyperviscosity syndrome.
Raynaud phenomenon
Cerebral venous thrombosis hyperviscosity syndrome
Impaired vision (e.g., blurry vision) The large size of IgM leads to
increased blood viscosity.
Retinal haemorrhages, engorged retinal veins
Headaches
Myeloproliferative neoplasms (MPNs)
A group of disorders characterized by a proliferation of normally developed
(non dysplastic) multipotent hematopoietic stem cells from the myeloid cell line.
According to the WHO classification, the following disorders are
myeloproliferative neoplasms:
Common (classic) Less common
Polycythemia vera Essential thrombocythemia
Primary myelofibrosis Chronic eosinophilic leukemia
Chronic myeloid leukemia Chronic neutrophilic leukemia
Myeloproliferative neoplasms, unclassifiable
Chronic myeloid leukemia (CML)
Is a type of myeloproliferative neoplasm involving hematopoietic stem cells
that results in overexpression of cells of myeloid lineage, especially
granulocytes.
Epidemiology
♂>♀
Peak incidence is 50–60 years.
Etiology
Idiopathic (in most cases)
Ionizing radiation (e.g., secondary to therapeutic radiation)
Aromatic hydrocarbons (especially benzene).
Pathophysiology
Philadelphia chromosome
Reciprocal translocation between chromosome 9 and chromosome 22 →
formation of the Philadelphia chromosome t(9;22) → fusion of the ABL1 gene
(chromosome 9) with the BCR gene (chromosome 22) → formation of the BCR-
ABL gene → encodes a BCR-ABL non-receptor tyrosine kinase with increased
enzyme activity.
Result: inhibits physiologic apoptosis and increases mitotic rate →
uncontrolled proliferation of functional granulocytes.
Clinical features
1) Chronic phase
2) Accelerated phase
Blast crisis
The blast crisis is the terminal stage of CML.
• Symptoms resemble those of acute leukemia.
• Rapid progression of bone marrow failure → pancytopenia, bone pain
• Severe malaise
Diagnostics
3) Diagnostic confirmation:
Flow cytometry:
Can be used to assess the type and maturity of leukocytes in order to detect progression
to advanced phases of CML.
Primary myelofibrosis
Bone marrow fibrosis, extramedullary hematopoiesis, and splenomegaly
Pathophysiology:
genetic mutations → hyperplasia of atypical megakaryocytes → ↑ TGF-β → ↑
fibroblast activity → bone marrow obliteration due to fibrosis → displacement of
hematopoietic stem cells → extramedullary hematopoiesis
Clinical features
Anemia
Symptomatic splenomegaly
Thromboembolic events
Petechial bleeding
Increased infections
Diagnostics of Primary myelofibrosis
CBC Thrombocytosis, and leukocytosis
↑ Leukocyte alkaline phosphatase, ↑ LDH
Peripheral blood smear: dacrocytes (teardrop cells)
Genetic marker JAK2 mutation
Bone marrow Aspiration Aspiration often fails (dry tap) because of severe marrow fibrosis.
Essential thrombocythemia
Isolated uncontrolled proliferation of platelets not caused by another condition
(e.g., reactive thrombocytosis, another myeloproliferative neoplasms).
Pathophysiology:
Genetic mutations → activation of the thrombopoietin receptor → proliferation of
platelets
Clinical features
• Increased risk of fetal loss
• Vasomotor symptoms (headache, visual disturbances, acral paresthesias,
ocular migraines)
• Erythromelalgia
• Acute gouty arthritis
• Thromboembolic events
• Petechial bleeding
• Commonly asymptomatic
Erythromelalgia
Diagnostics
Pathophysiology
The JAK2 (Janus kinase 2) oncogene codes for a non-receptor tyrosine
kinase in hematopoietic progenitor cells.
JAK2 is essential for the regulation of erythropoiesis,
thrombopoiesis (megakaryopoiesis), and granulopoiesis.
95% of primary PV patients have a mutation in the JAK2 gene (gain of
function) → ↑ tyrosine kinase activity → erythropoietin-independent
proliferation of the myeloid cell lines → ↑ blood cell mass (erythrocytosis,
thrombocytosis, and granulocytosis) → hyperviscosity and slow blood flow
→ ↑ risk of thrombosis and poor oxygenation.
Clinical features
Often asymptomatic
Hyperviscosity syndrome (triad of mucosal bleeding, neurological symptoms, and visual changes)
Plethora (Plethora)
Cyanotic lips
Pruritus; worsens when the skin comes into contact with warm water
neurological symptoms: dizziness, headache, visual disturbances, tinnitus
Hypertension
Splenomegaly
Peptic ulcer disease
Symptoms of thrombotic and hemorrhagic complications.
Diagnostics
Increase Hb/Hct, RBCs, Platelets and Leukocytes (> 12,000/μL)
Decrease ESR and EPO
Etiology
Treatment-related MDS:
Following cytostatic therapy (alkylating agents, topoisomerase II inhibitors,
azathioprine, etc.)
Clinical features
1) Hepatosplenomegaly
2) Depending on the affected cell line:
Erythrocytopenia; anemia
Leukocytopenia increased susceptibility to bacterial infections, especially of the skin
Thrombocytopenia; petechial bleeding
Diagnostics
Ringed sideroblasts:
Haematinics:
Haematinics are nutrients required for the formation of blood cells. Iron, vitamin
B12, Erythropoietin and folate are the main haematinics, and deficiencies can
lead to anaemia.
Patients with a 𝑯𝒃 𝒍𝒆𝒔𝒔 𝒕𝒉𝒂𝒏 𝟕𝒈/𝒅𝒍 will have symptoms of tissue hypoxia
(fatigue, headache, dyspnea, pallor, angina, tachycardia, visual impairment,
syncope, lymphadenopathy (enlargement of lymph nodes), hepatic and or splenic
enlargement, bone tenderness, blood loss in feces, neurologic symptoms.
In addition to general signs and symptoms of anemia, iron deficiency anemia
may cause:
Pica Hunger for ice. dirt, paper, etc.
Koilonychias Upward curvature of the finger and toe nails (spoon nail).
Angular cheilitis Soreness and cracking at the comers of the mouth.
1) Oral iron:
Drug Volume of drug elemental iron
Ferrous sulfate 325 𝒎𝒈 65 𝒎𝒈
Ferrous gluconate 320 𝒎𝒈 37 𝒎𝒈
Ferrous fumarate 325 𝒎𝒈 106 𝒎𝒈
Ferric iron binds with transferrin in plasma and transported in other tissues
and stored as ferritin and hemosiderin form.
This complex bind with receptor on developing red cells in B. marrow, iron
released in the cell, transferrin and transferrin receptor are then recycled,
providing an efficient mechanism for incorporating iron into hemoglobin in
developing cells.
Storage: Iron storage as Ferritin and hemosiderin form in mucosal cells, liver,
spleen, and bone marrow.
Hemosiderin large particles can be observed microscopically
ferritin So small particles Can be seen in the cell cytoplasm only
with an electron microscope.
Elimination: Minimal amount (about 1 mg/day) are lost in sweat, saliva, and in
exfoliated skin and intestinal mucosal cell.
man woman
An average excretes about 0.6 mg of iron Additional menstrual loss of
each day, mainly into the feces. Additional blood brings long-term iron loss
quantities of iron are lost when bleeding to an average of about 1.3
occurs mg/day.
Iron deficiency anaemia is the only indication for the use of iron.
The most common cause of iron deficiency anaemia in adults is blood loss.
200- 400mg oral elemental iron daily should be given to correct anaemia (25%
absorbed, so 50-100mg iron can be incorporated in Hb).
Treatment should be continue for 3-6 months, this not only correct the anemia but
will replenish iron stores. (Hb should reach normal level in 1-3 months).
Failure to respond to oral iron therapy may be due to incorrect diagnosis.
Indications:
1. It should be reserved for patients with documented iron deficiency unable to tolerate
or absorb iron (pts. With post gastrectomy, previous small bowel resection,
malabsorption syndrome).
2. Pts. With extensive chronic blood loss who cannot be maintained with oral iron alone.
Iron dextran:
Combination of ferric hydroxide + dextran
50 mg elemental iron/ml
Route of administration: IM, or by IV infusion in 1-2 hours.
Most adults needs about 1-2 G (20-40ml) iron dextran for iron deficiency
anemia.
It is seen in young children who have ingested a no. of iron tablets (more than 10
tablets).
Adults are able to tolerate large doses of iron.
2) Local pain, tissue staining (brown discoloration of tissues overlying the inj. site),
headache, fever, arthralgia, nausea, vomiting, bronchospasm, urticaria, anaphylaxis,
and death.
Treatment of acute iron toxicity:
1) Whole bowel irrigation should perform.
2) Deferoxamine {iron antidote}; is a potent iron chelating.
3) Supportive therapy for GIT bleeding, metabolic acidosis and shock.
Treatment of hemochromatosis:
1) Intermittent phlebotomy, 1 unit of blood removed weekly.
2) Iron chelating agent (deferoxamine IV). Phlebotomy – 350 ml
Transfusion – 500 ml
Deferoxamine:
Is a potent iron chelating; binds iron that has already been absorbed and to
promote its excretion in urine and feces.
It is poorly absorbed when given orally and may increase iron absorption by this
route.
It is given IM or preferably IV.
It is metabolized and excreted in urine (turn urine color orange red).
في مادة معينة باأللفيوالي سائلة تتصنع بشكل دائم الزمة لمنع ايديما الرئة
ونقص هذه المادة يؤدي الى هذا المرض
Megaloblastic anemias:
are a group of disorders characterized by defective nuclear maturation caused
impaired DNA synthesis, caused usually due to vitamin B12 or folate deficiencies.
Vitamin B12:
Water soluble with MW 1335 Daltons.
Produced by micro-organisms and fungi.
The recommended dietary intake for adult is 2µg/day.
In this reaction both vit B12 and folic acid are involved.
B12 acts as a co-enzyme (methyl cobalamin) for methyltransferase.
The use of folic acid alone in the presence of vitamin B12 deficiency may result
in worsening of neurological defects.
Vitamin B12 deficiency Folate deficiency
Vitamin B12 deficiency is treated with Oral folic acid 5 mg daily for 3 weeks
hydroxycobalamin 1000 𝝁𝒈 (𝟏𝒎𝒈). will treat acute deficiency and 5 mg once
weekly is adequate for maintenance
Five doses 2 or 3 days (or 10 days) therapy.
apart followed by maintenance
therapy of 1000 𝝁𝒈 every 3 months for Prophylactic folic acid in pregnancy will
life. prevent megaloblastosis in women at risk.
1) Hydroxyurea:
Effective in reducing painful episodes by about 50%.
The necessity of blood transfusions was also shown to be reduced.
Hydroxyurea increases the production of fetal hemoglobin, which makes red
cells resistant to sickling.
The goal of sickle cell anemia drugs is to vasodilation, but Hydroxyurea alone cannot
do this, so we must give it a second drug "Pentoxifylline".
Treatment of Sickle cell anemias “continue”
2) Pentoxifylline:
Is a synthetic dimethyxanthine structurally similar to caffeine.
The actions of pentoxifylline include increased erythrocyte flexibility and
decreased blood viscosity.
It is commonly used to treat intermittent claudication ()العرج, by improving blood
flow and tissue oxygenation.
Pentoxifylline appears to inhibit erythrocyte phosphodiesterase, which causes
an increase in erythrocyte cyclic adenosine 5ʹ-monophosphate activity and an
increase in membrane flexibility.
Hematopoieti
c factors
Erythrocyte Granulocyte
platelet
factors factors
factors
Oprelvekin
vitamins Iron Erythropoiesis- Filgrastim Saigramostim
stimulating agents (IL-11) (G-CSF) (GM-CSF)
(B12, B9)
(ESA)
Darbepoetin and
erythropoietin
Hematopoietic growth factors
Are endogenous glycoproteins that bind to specific receptors on bone marrow
progenitor cells and induce their differentiation and proliferation, thereby increasing
production of erythrocytes and various leukocytes.
Several growth factors are now available for treating anemia or leukopenia.
These growth factors are produced by recombinant DNA technology and are
administered parenterally.
The endogenous forms of these growth factors are produced by various leukocytes,
fibroblasts, and endothelial cells.
The addition of a PEG moiety to filgrastim (pegylation) creates pegfilgrastim, whose
molecular size is too large to enable renal clearance, thereby increasing the half-life from
about 3.5 hours for filgrastim to 42 hours for pegfilgrastim.
Adverse effects:
The most common adverse effects of IL-11 are fatigue, headache, dizziness,
and fluid retention.
Thrombopoietin agonist Romiplostim: Eltrombopag:
Rout of administration SC receptor agonist Oral agonist
Chronic idiopathic Chronic idiopathic
Indications thrombocytopenia who thrombocytopenia that is
have failed to respond to refractory to other
conventional treatment. agents.
Limitation of use Unrestricted restricted
Risks ---------------------------------- Hepatotoxicity and
hemorrhage.
Chloramphenicol cause:
Aplastic anemia.
Note that both warfarin and heparin affect in common pathway (X+ II).
3 stages of hemostasis in order:
1. Vascular spasm. Evaluated by bleeding time
2. Formation of a platelet plug.
3. Blood coagulation.
The first two stages enough to stop bleeding in small vascular injury.
The blood vessel spasm refer to some mechanisms;
Platelet substances: ADP, TXA2, histamine and serotonin.
Nervous reflex.
Vascular smooth muscles tune.
Notes:
Active plasmin degrade fibrin, fibrinogen.
Some drugs and substances activate plasminogen into plasmin:
Rivaroxaban inhibit the activity of Stuart factor (factor Xa) and thrombin
Drugs Affecting Clot Formation and Resolution:
Interfere with the clotting cascade and thrombin
Anticoagulants formation. Heparin, Warfarin
Prevent new thrombus to form.
Antiplatelets Alter the formation of the platelet plug Aspirin, Clopidogrel
{Disaggregation of platelets}.
Thrombolytic Break down the thrombus that has been formed by Alteplase,
drugs stimulating the plasmin system. Urokinase,
Strentokinase
Mechanism of action:
Heparin acts indirectly by binding to antithrombin III, causing a rapid anticoagulant effect, in vivo
and vitro, effect occurs within minutes after I.V injection, in the absence of heparin, antithrombin III
interacts very slowly with thrombin and factor Xa. Heparin molecules bind antithrombin III inducing a
conformational change that accelerates its rate of action about 1000-fold.
Heparin serves as a true catalyst, allowing antithrombin III to rapidly combine with and inhibit
circulating thrombin and factor Xa, antithrombin III inhibits II, IX, X, XI, and XII
Chemistry of Heparin:
Absorption:
Heparin is poorly absorbed from the gut, because of its charged and large molecules, so it
must be given parenterally, either by deep S/C (effects appear in 1-2 hours after injection) or
I.V.
I.M injection is contraindicated, because of the formation of hematoma.
Orally contraindicated also.
Heparin rapidly bind to plasma proteins inactivated in the liver and excreted in the urine;
diseases of liver and kidney prolong its t 1/2.
Placental barrier:
Therapeutic Uses:
a. Deep vein thrombosis and pulmonary embolism: decreases the incidence of recurrent
thromboembolism.
b. Prophylaxis of postoperative venous thrombosis in patients undergoing elective surgery
(hip replacement) and those in the acute phase of MI.
C. Pregnant women with thromboembolism or with prosthetic heart valves.
d. The drug is also used in extracorporeal devices (dialysis machines) to prevent thrombosis.
Heparin Toxicity:
Bleeding; is the major side effect of heparin.
Heparin is monitored by two ways;
1) Anti-Xa units (0.2-0.7 units).
2) Activated partial thromboblastin time (aPTT): defined as the time required for plasma to
clot in the presence of kaolin (activator of XII), cephalin (substitute PL) and Ca, normally
For example: In heparin toxicity, every 100 mg of protamine sulfate is given. If the original dose was
1000 mg of heparin, we do not give 10 mg of protamine, but we measure the concentration of the drug
in the plasma when toxicity occurs, and suppose that it is 600 mg, we give + mg of protamine.
Fondaparinux:
Exert their anticoagulant effect by directly binding to the active site of thrombin (inhibition)
in contrast to indirect thrombin inhibitors as LMWHs which act through binding (activation)to
antithrombin.
Clinical Use:
4. Toxicity:
Like other anticoagulants, the factor Xa inhibitors can cause bleeding.
Pharmacokinetics:
Warfarin is well absorbed, following oral administration has a high
bioavailability, 99 % of warfarin bind to plasma albumin, which prevents its
diffusion into the CSF, urine and breast milk, drugs that have a greater affinity for
albumin binding site, such as sulfonamides, can displace the anticoagulant and
lead to a transient elevated activity.
It has a low volume of distribution, but a long t1/2 36 hours, metabolized in the
liver into inactive metabolites that are excreted in the urine and stool.
Warfarin does cross the PBB.
Mechanism of action:
Normally vitamin K is involved in the synthesis of active prothrombin and other
clotting factors (VII, IX and X), this appears in conversion of glutamic acid to y-
carboxyglutamic acid in the presence of O2, CO2 and active form of vitamin K
(KH2) (hydroquinone).
The above reaction requires reactivation of vitamin K, that occurs by oxidation
of vitamin K epoxide (KO) into its active form via vitamin K epoxide reductase
enzyme (VKER).
VKER is inhibited by warfarin, the result is inactive biologically
(noncoagulatory) proteins, in addition to this action warfarin activates
physiologic anticoagulants especially C and S proteins.
Sources of Vitamin K:
K1 K2 K3
phytonadione menaquinone Menadione
green leafy vegetables gut flora synthetic
Fat soluble Fat soluble water soluble
Properties of Warfarin:
1) It is active only in Vivo.
2) Onset of anticoagulant effect is only after 8 to 12 hours; maintenance dose is
achieved in 24 hours, even if it is given I.V (rare).
3) The anticoagulant effect persists for a few days, after the drug has been
stopped.
Prothrombin time (PT): is the time required for plasma to clot in the presence of
exogenous thromboplastin, the normal PT is 11-15 seconds.
high INR (ex; 5) Low INR (ex; 0.5)
high chance of bleeding high chance of having a clot
Adverse Effects of warfarin:
a. Bleeding disorder:
Is the most common side effect, especially hemorrhage, of the bowel or the brain,
which is reversed by administration of vitamin K1 and fresh plasma.
However, reversal following administration of vitamin K takes
approximately 24 hours.
b. Fetus toxicity:
In addition to bleeding there is a high risk of abnormal bone formation, because
warfarin prevents the formation of y-carboxyglutamic acid, which found in the
bone, so it should be avoided in pregnancy.
c. Hepatitis and necrosis of soft tissues are rare symptoms.
Bilirubin may displace warfarin, causing Kernicterus
VWF functions;
1) By GP Ilb/IIIa forms bridges between platelets and subendthelium.
2) Carrier of VIII clotting factor.
Clinical Uses:
These agents are beneficial in the prevention and treatment of occlusive
cerebrovascular, peripheral vascular disease {PVD} and cardiovascular diseases,
in the maintenance of vascular grafts and arterial patency, and as adjuncts to
thrombin inhibitors or thrombolytic therapy in myocardial infarction.
Prostaglandin Thromboxane A2
Vasodilation. Vasoconstriction.
Inhibit platelet aggregation. Activate platelet aggregation.
Antiplatelet drugs:
A. Aspirin:
Is the only irreversible COX inhibitor that prevents TXA2 synthesis, the
inhibitory effect is rapid, apparently occurring in the portal circulation.
Aspirin is frequently used in combination with other drugs having anticlotting
properties for example, heparin or clopidogrel.
NSAIDs, such as ibuprofen, inhibit COX-1 by transiently competing at the
catalytic site.
Ibuprofen, if taken concomitantly ( )في نفس الوقتwith, or 2 hours prior to
aspirin, can antagonize the platelet inhibition by aspirin.
Therefore, aspirin should be taken at least 30 minutes before ibuprofen or at
least 8 hours after ibuprofen.
C. Abciximab:
is a monoclonal antibody, that binds to GPIlb/IIIa receptors, the antibody
blocks the binding of fibrinogen and von Willebrand factor; consequently,
aggregation does not occur, another member of this group is eptifibatide.
E. Dipyridamole:
A coronary vasodilator is employed prophylactically for angina pectoris.
It is usually given in combination with aspirin or warfarin; it is ineffective when
used alone.
Dipyridamole increases intracellular levels of cAMP by inhibiting cyclic
nucleotide phosphodiesterase, resulting in decreased thromboxane A2 synthesis.
It may potentiate the effect of prostacyclin to antagonize platelet stickiness
and, therefore, decrease platelet adhesion to thrombogenic surfaces.
Dipyridamole is effective for inhibiting embolization from prosthetic heart
valves.
Thrombolytic Agents
Mechanism of Action:
Activate plasminogen to plasmin, which in turn breaks down fibrin threads in a
clot to dissolve a formed clot.
Indications:
Acute MI, severe pulmonary emboli, DVT and to clear occluded venous catheters.
Contraindications:
Allergy.
Any condition that worsens through dissolution of clots.
Pregnancy and lactation.
Alteplase and Reteplase bind directly with fibrin and lysis it.
Not activate plasmin.
Streptokinase and Urokinase activate plasmin directly.
1) Cardiac arrhythmias.
2) Hypotension.
3) Bleeding:
Is the most important hazard and has about the same frequency with all the
thrombolytic drugs.
Cerebral hemorrhage is the most serious manifestation.
Bleeding of GI, genitourinary, respiratory and retroperitoneal also occur.
4) Hypersensitivity reactions:
Rash, flushing ()توهج, bronchospasm, and anaphylactic reaction, specifically for
Streptokinase.
Streptokinase; a bacterial protein, can evoke the production of antibodies that
cause it to lose its effectiveness or induce severe allergic reactions on
subsequent therapy.
Patients who have had streptococcal infections may have preformed
antibodies to the drug.
Bleeding Disorders Treated With Clotting Factors
1) Hemophilia:
Genetic lack of clotting factors that leaves the patient susceptible to excessive
bleeding from any injury.
2) Liver disease:
Clotting factors and proteins needed for clotting are not produced.
Vitamin K.
Plasma fractions.
Fibrinolytic inhibitors aminocaproic acid.
Serine protease inhibitors aprotinin.
HEMOSTATIC DRUGS
1. Vitamin K:
Is a fat-soluble vitamin, which requires bile salts for absorption form the gut.
Pharmacokinetics:
tablets ampules
5mg 50 mg
The onset of action is delayed for 6 hrs and accomplished by 24 hrs.
Clinical Indications of vitamin K:
1. Warfarin toxicity: infusion should be done slowly, because it can cause
dyspnea, chest and back pain leading even to death.
2. Vitamin K deficiency: as supplemental therapy either due to disorders of liver,
intestine diseases, or in hospitalized patients in ICU because of poor diet,
parenteral nutrition, recent surgery, multiple antibiotic therapy and uremia.
3. Vitamin K is currently administered to all newborns to prevent hemorrhage
disease of vitamin K deficiency, which is especially common in premature
infants.
2. Plasma Fractions:
A) Plasma factors:
Freeze concentrates of plasma containing prothrombin, factors IX and X and VII
are commonly available for treating deficiencies of these factors e.g. hemophilia
B is treated by IX factor.
B) Desmopressin Acetate:
Increases activity of VIII factor and used to treat hemophilia A (deficiency of VIII
factor or von-Willebrand disease.
Desmopressin acetate can be used in preparation for minor surgery such as
tooth extraction without any requirement for infusion of clotting factors if the
patient has a documented adequate response.
c. Cryoprecipitate:
Plasma protein fraction obtained by whole blood; used to treat deficiencies of
fibrinogen such as that occurs in DICS and liver disease, VIII deficiency and von-
Willbrand disease if desmoprosin is not indicated.
Mechanism of action:
Inhibit competitively fibrinolysis (conversion of plasminogen into plasmin or
fibrinolysin) thus preventing lysis of fibrin.
Pharmacokinetics:
The drugs are rapidly absorbed orally, and cleared from the body via the kidneys,
administered IV as loading dose of 5 g, over 30 minutes to avoid hypotension.
Clinical Uses:
They are used as adjunctive therapy ( )كعالج مساعدin hempophilia, bleeding form
fibrinolytics, (streptokinase, urokinase and alteplase), prophylaxis for rebleeding
from intracranial aneurysms, post GIT bleeding, hemorrhage of the urinary bladder,
secondary to radiation- and drug-induced cystitis and postprostatoectomy.
Adverse Effects:
Intravascular thrombosis, hypotension, diarrhea, myopathy, nasal stuffiness and
abdominal discomfort.
Contraindications:
DICS and genitourinary bleeding from the upper tract because of high incidence of
excessive clotting.
Adverse effects:
Cardiac arrhythmias, MI, CHF, hypotension, increased risk of renal failure, heart
attack, and stroke.
Antineoplastic Drugs:
Cancer:
Is a term used for diseases in which abnormal cells divide without control and
are able to invade other tissues by metastasis.
Nomenclature;
In general benign tumor attaching with the suffix (- oma).
Epithelial Mesenchymal
Adenoma Lipoma
Papilloma Fibroma
Liquid cancers are not treated with surgery, but with chemotherapy, unlike solid cancers,
which require surgery before chemotherapy.
The most common organs in which cancer has spread, in order:
Liver lung brain bone
Anticancer drugs; {A-A-A-C-M-T-V}
A – A – A:
A A A
ALKYLATING AGENTS ANTIMETABOLITES ANTIBIOTICS
1) Usually start with {c}. Usually end with: Usually end with;
2) May end with: ate. mycin.
mide. purine. bicin.
fan. bine.
lan Xed.
amine Remember; Mitoxantrone
ine Remember; 5-Fluorouracil
tin
C – M – T – V:
CAMPOTHECIN ANALOGUES MISCELLANEOUS TAXANES VINCA ALKALOIDS
Ends with: No uniform rule Ends with: start with:
tecan taxel Vin
1) Interphase;
2) Mitotic phase:
Prophase.
Metaphase.
Anaphase.
Telophase.
After completion of mitosis, the resulting daughter cells have two options:
They can either enter G1& repeat the cycle.
They can go into G0 and not participate in the cell cycle.
Growth fraction
The ratio of proliferating cells to cells in G0.
large percentage of proliferating cells & few cells in G0 mostly of cells in G0
high growth fraction low growth fraction
People being treated with chemotherapy are given filgrastim and pegfilgrastim.
According to their actions on the cell cycle, antitumor agents are divided into:
Cell Cycle-Specific (CCS) Agents: Cell- Cycle Non-Specific (CCNS) Agents:
effective for high growth-fraction effective for both low growth fraction
tumors, (only against replicating cells), tumor (solid) as well as for high growth
Affect specific phase in cycling cells can kill both Go and cycling cells
antimetablities, antitumor antibiotics Alkylating agents, antitumor antibiotics
(bleomycin), vinca alkoloids, (dactinomycin and mitomycin)
etoposide and taxanes. anthracyclines, camptothecins and
platinum analogs.
PALLIATIVE CHEMOTHERAPY:
Initial remissions are transient, with symptoms recurring between treatments.
Survival is extended, but the patient eventually dies of the disease.
Principles of Cancer Chemotherapy:
Tumor susceptibility and the growth cycle:
Rapidly dividing cells are generally more sensitive to anticancer drugs where as
non-proliferating cells (those in phase Go) usually survive the toxic effects of
these agents.
The growth rate of tumor initially is rapid, but decreases as the tumor size
increases due to inadequate vascularization.
Reducing the tumor burden through surgery or radiation, increases their
susceptibility to anticancer drugs.
Pulse therapy:
Involves intermittent treatment with very high doses of anticancers that are too
toxic to be used continuously.
This method is used successfully in therapy of acute leukemias and testicular
carcinomas.
Rescue therapy:
Some neoplastic cells are inherently resistant to most anticancer drugs, (e.g.
malignant melanoma renal and brain cancers absence of response on the first
exposure), properly due to mutation of p 53 suppressor (decreased its activity),
gene resulting in resistance to radiation therapy and to a wide range of anticancer
drugs.
Acquired resistance:
Their maximal cytotoxic effects are in S-phase and are, therefore, cell cycle
specific.
Methotrexate (MTX)
MTX is structurally related to folic acid (vitamin B9) and act as antagonist of it.
It inhibits mammalian dihydrofolate reductase (DHFR).
The inhibition of DHFR can only be reversed by:
1000-fold excess of the natural substrate, dihydrofolate (FH2).
By administration of leucovorin, which bypasses the blocked enzyme and
replenishes the folate pool {rescue therapy}.
M.O;
6-MP and 6-TG are activated by hypoxantheine-
guanine phosphororibosyltransferases (HGPRTases)
to form 6- mercaptopurine-ribose phosphate (6-MPRP)
and other toxic metabolites (like; TIMP).
(6-MPRP) and other toxic metabolites (like; TIMP);
inhibit several enzymes of de novo purine nucleoside
synthesis.
Resistance:
Is usually due to decreased conversion to F-UMP.
Pharmacologic properties:
5-Fluorouracil is administered IV; it is also used topically to treat skin cancers.
Vinca alkaloids
VinBlastine and VinCristine (oncovan)
VinBlastine VinCristine
Uses ABVD MOPP
A Adriamycin (doxorubicin) M Mechlorethamine
State B Bleomycin O Vincristine (Oncovin),
V Vinblastine P Procarbazine
D Dacarbazine P Prednisone
Hodgkin’s disease. Childhood leukemias.
INDICATION Lymphomas Breast carcinoma.
Childhood tumors-Wilm’s tumor.
(Neuroblastoma).
Testicular tumors
Hodgkin’s disease.
Bone marrow suppression. Peripheral neuritis.
TOXICITY Anorexia. Paresthesia.
Nausea, vomiting. & Diarrhea. Muscle weakness.
Alopecia
VII. ADJUNCT AGENTS:
Amifostine (Ethyol):
Is a cytoprotective agent that is dephosphorylated to active-free thiol, which then acts
as a scavenger of free radicals.
It is used to:
1) Reduce the incidence of neutropenia-related fever.
2) Reduce infection induced by alkylating agents and platinum-containing agents (e.g.,
cisplatin.)
3) Reduce renal toxicity associated with platinum-based drug therapy.
4) Reduce xerostoma (sensation of dry mouth) in patients undergoing irradiation of
head and neck regions.
Characteristics of “Important” Anticancer Drugs
1) Alkylating agents: non-specific cell cycle
Drugs Mechanism Used Adverse effect
Non-Hodgkin’s, 1) BMS.
Cyclophosphamide attacks guanine ovarian, breast CA, 2) Mucositis.
N7— dysfunctional neuroblastoma 3) Hemorrhagic cystitis.
DNA 4) hepatotoxicity (high
dose)
Testicular. 1) Nausea, vomiting
Ovarian. (use ondansetron)
Cisplatin cross links DNA Bladder. 2) Nephrotoxicity
strands lung CA. (use amifostine)
3) Neurotoxicity
(use deafness)
1) BMS.
Procarbazine 2) Pulmonary toxicity,
Hodgkin’s (MOPP) 3) hemolysis.
4) Neurotoxicity.
5) Leukemogenic.
IMMUNE MECHANISMS:
Cell –mediated immunity Humoral (antibody –mediated
immunity).
T- lymphocytes B- lymphocytes
Immune potentiators
Aldesleukin Interferons
Cytokines:
Soluble, antigen-nonspecific signaling proteins
that bind to cell surface receptors on a variety of
cells.
Cytokines include;
Interleukins.
Interferons (IFNs).
Tumor Necrosis Factors (TNFs).
Transforming Growth Factors (TGFs).
Colony-stimulating factors (CSFs).
Uses of Cyclosporine:
Indications:
Organ and stem cell transplantation, prevention of rejection of liver and kidney
transplants and atopic dermatitis and psoriasis (topically).
Toxic effects:
Anaphylaxis.
GIT disturbances.
NO hirsutism or gum hyperplasia.
Drug interactions as cyclosporine.
Hyper (glycaemia, tension and kalemia).
Nephrotoxicity (more than CsA).
Neurotoxicity (more than CsA).
Anti-inflammatory action:
Immunosuppressant action:
Pharmacokinetics:
Orally or intravenously.
Widely distributed but does not cross BBB.
Metabolized in the liver to thiouric acid (inactive metabolite) by xanthine
oxidase and excreted primarily in urine.
Drug Interactions:
Co-administration of allopurinol with azathioprine may lead to toxicity due to
inhibition of xanthine oxidase by allopurinol.
USES:
Acute glomerulonephritis, Systemic lupus erythematosus, Rheumatoid
arthritis, Crohn’ s disease and autoimmune hemolytic anemia.
Adverse Effects:
Bone marrow depression: leukopenia, thrombocytopenia,
GIT toxicity, Hepatic dysfunction and Increased risk of infections.
MYCOPHENOLATE MOFETIL
Chemistry:
Is a semisynthetic derivative of mycophenolic acid from fungus source.
Metabolism:
Is a prodrug, hydrolyzed to mycophenolic acid, it is extensively bound to
plasma protein, metabolized in the liver by glucuronidation and excreted in urine.
Mechanism of action:
Inhibits de novo synthesis of purines.
Mycophenolic acid is a potent inhibitor of inosine monophosphate (IMP),
crucial for purine synthesis which leads to deprivation of proliferating T and B
cells of nucleic acids.
CLINICAL USES:
In solid organ transplantation.
Hematopoietic stem cell transplant patients.
Combined with tacrolimus as prophylaxis to prevent graft versus host
disease.
In autoimmune disorders: Rheumatoid arthritis, & dermatologic disorders.
Sulfapyridine Anti-bacterial
Peptic ulcer and chron
5-ASA Anti-inflammatory
disease
Antibodies:
Sometimes used as a quick and potent immunosuppressive therapy to prevent
the acute rejection reactions.
Polyclonal antibodies: Monoclonal antibodies:
Antilymphocyte globulins (ALG). Basiliximab.
Antilymphocyte globulins (ALG). Daclizumab.
Polyclonal antibodies:
Bind to the surface of circulating T lymphocytes, which are phagocytosed in the
liver and spleen giving lymphopenia and impaired T-cell responses & cellular
immunity.
Kinetics:
Given i.m. or slowly infused intravenously.
Half-life extends from 3-9 days.
Adverse Effects:
Antigenicity.
Anaphylactic and serum sickness reactions (Fever, Chills, Flu-like syndrome).
Leukopenia, thrombocytopenia.
Risk of viral infection.
Monoclonal antibodies:
Are IL-2 receptor antagonists, Bind to CD25, Block IL-2 stimulated T cells
replication & T-cell response system.
Given I.V.
Drug Basiliximab Daclizumab
Potential More potential Less potential
Half life 7 days 20 days
Infliximab
Is targeted against TNF-α, a proinflammatory cytokine, and thereby decreases
formation of interleukins and adhesion molecules involved in leukocyte
activation.
Infliximab induces remissions in treatment-resistant Crohn’s disease.
In combination with methotrexate, infliximab improves symptoms in patients
with rheumatoid arthritis.
It also is effective in the treatment of ulcerative colitis, ankylosing spondylitis,
and psoriatic arthritis.
Infusion reactions and an increased rate of infection may occur.
Etanercept:
Is not a true Mab, binds with high affinity to TNF-α.
Etanercept is used in arthritis, psoriasis, and ankylosing spondylitis, and it is
being investigated in other inflammatory diseases.
1. Salmonella
Gram negative bacilli, Facultative anaerobic.
Belong to Enterobacteriaceae (Gammaproteobacteria).
Motile by peritrichous flagella (distributed over the entire cell).
Tolerate stomach acidic pH.
Live in intestines of all vertebrates.
Not part of human normal microbiome {Potential pathogen}.
Encapsulated bacteria.
Contain endotoxin.
Contain O antigen.
Contains pili which encoded by plasmids.
Caused osteomyelitis (for sickle cell disease patients).
Positive for hydrogen sulfate (H2S)
Classification of Salmonella:
Antigenic variation.
Thermostable.
2) H antigen:
A. Phase I H antigens:
Responsible for immunological identity expressed by some serotypes.
B. Phase II antigens:
Non-specific antigens, found in many other serotypes.
3) Vi (virulence) = capsule:
4) Salmonella Pathogenisity
Fluoroquinolones
Prevention:
1) Vaccination: live attenuated mutant strain of Salmonella Typhi, Vi capsular
polysaccharidevaccine.
2) Improve hygiene and sanitation.
2. Brucella
Aerobic, gram- negative coccobacilli.
Belong alphaprotobacteria.
Facultative intracellular.
Noncapsulated.
Non motile.
Cause zoonotic infection called brucellosis (malta fever, undulent fever).
In animal hosts, the Brucella lives as an intracellular parasite in organs such as the
uterus and placenta.
It has been proposed that there is only B. melitensis based on DNA-DNA
hybridization.
Some studies depend that B. melitensis is the only species, but there are
many species, the most important of which are: B. abortus and B. suis,
where the most dangerous is B. melitensis.
Inhibits:
Phagosomal fusion and lysosomal destruction → allows
Lipopolysaccharide intracellular survival.
(LPS) Complement deposition (opsonization).
Endotoxin:
Less toxic than in other species of bacteria.
Weak immune system inducer.
Superoxide Provides defense against oxidative burst
dismutase and activity → allows intracellular survival.
catalase:
Type IV secretion Multi-protein complex.
system: Allows Brucella to reach and replicate within a cell’s
endoplasmic reticulum.
Transmission of Brucella
Ingestion Spoil milk
Direct contact Touching the wool ) (صوفof an infected
animal.
Inhalation Dust from wool or other dried material
of infected animals.
Accidental inoculation Laboratory workers who handle
culture of the organism.
Transplantation and blood transfusion Very rare.
Diagnosis of Brucella:
1) Culture:
Definitive.
Done in the first week.
Shows positive within 3-7 days.
Longer incubation (up to 6 weeks) may be needed.
Bone marrow is the gold standard (most sensitive).
Requires special culture techniques.
The prevention:
Pasteurization of dairy products.
Immunization of uninfected domesticated animals.
Proper disinfection of urea and removal of reproductive tissues.
Slaughter { }ذبحof infected animals.
Plasmodium:
Obligate intracellular parasite belong
apicomplexan group.
The Apicomplexa
Are not motile in their mature forms
Non motile.
and are obligate intracellular
Contain enzymes that penetrate the host’s parasites.
tissues at the apical area. They’re characterized by formation
of oocyst.
Life cycle involves transmission between
Cantains apical complex, which
several hosts.
contains proteins facilate cell
Cause malaria, affects 10% of the world’s membrane pore “rhoptries and
population. micronemes”
In plasmodium
Definitive host Mosquito
Intermediate host Human
Plasmodium species:
Erythrocytes
P. Falciparum P. Vivax P. Ovale
Schuffner dots - + +
Maurer dots + - -
Parasites
P. Falciparum P. Vivax P. Ovale
All sexual stages seen - + +
Band forms - - -
Double infections + - -
Double chromatin dots + - -
Banana shaped gametocytes + - -
Note that (-) does not mean the absence of the property exclusively, but the (+)
confirms its existence very clearly; For example, Banana shaped gametocytes are
present at (P. Vivax and P. Ovale) but are more visible in (P. Falciparum).
Plasmodium pathophysiology
People with blood diseases such as sickle cell anemia, thalassemia or
hemoglobin C often tend to be less likely to get malaria because RBCs do not live
long.
There are many virulence factors of Plasmodium in the attack, and they are as
follows:
7) HLA-polymorphism:
9) Glucose-6-phosphate dehydrogenase:
Fever cycle:
After the RBCs burst, the plasmodium is released into the blood.
It is recognized by the macrophage and secretes IL-1, TNF-𝜶 , which is then activated by the hypothalamus; which raises
the body temperature.
Available Malaria Diagnostic Tests:
1) Clinical:
Anemia.
Increase liver enzyme expression, reticulocytes and jaundice.
Black water fever.
Dark urine.
Thrombocytopenia as a result of Rosetting formations; platelets life
span decrease.
3) RTDs:
We use it when the shape of the parasite is clear and easy to identify.
Rapid diagnosis {serological}; 2-lines and contain lactose filter paper.
When the parasite-laden blood binds to the antibody, the first line is
formed.
The fluid completes the movement until the formation of the second line.
Indication:
Clinical Microscopy and RTDs HS-RTDs and US molecular
molecular
Symptoms 100 – 200 𝒑𝒂𝒓𝒂𝒔𝒊𝒕𝒆𝒔 / 𝝁𝒍 2–10 <1
Management:
4) Vaccination:
Do not give lifelong immunity.
There is no study that proves that mice that have acquired the infection
transmit the infection to humans.
The infective part is the sporozoites but quickly turns into merozoites in
the mouse.
Life cycle:
The same life cycle of
Plasmodium, but the difference is
that it does not go through the liver
stage.
Babesia: Transmission
1) Transfusion of packed red cells (blood - plasma).
2) Rarely transplacental.
3) Reported in immunocompetent and immunocompromised individuals.
Immunocompetence is the ability of the body to produce a normal immune response following
exposure to an antigen.
Immunocompetence is the opposite of immunodeficiency or immuno-incompetent
or immuno-compromised.
2) Incubation period:
Depends on the route of transmission:
2) Molecular tests:
Highly sensitive and specific.
Useful for low parasitemia.
Confirm infection.
Determine species.
3) Serologic tests:
Indirect florescence antibody “IFA” and ELISA.
In endemic areas support or confirm the diagnosis of human babesiosis.
Useful in screening blood donors for Babesia infectns.
1) Bubonic plague
It's named after the swollen lymph nodes (buboes) that typically develop in the first week after
you become infected.
Possible bubonic plague signs and symptoms may include:
2) Septicemic plague
Septicemic plague occurs when plague bacteria multiply in your bloodstream.
Possible Septicemic plague signs and symptoms may include:
Nausea, vomiting, diarrhea, and abdominal pain.
Disseminated intravascular coagulation (DIC).
Gangrene in the limbs may turn black.
Organs malfunction (lung, spleen).
Death if not treated.
3) Pneumonic plague
Pneumonic plague affects the lungs. It's the least common variety of plague but the most
dangerous.
Signs and symptoms can begin within a few hours after infection, and may include:
Fever.
Headache.
Weakness.
Cough.
Hemoptysis.
Dyspnea.
Chest pain.
Yersinia pestis life cycle:
We put the mucus on a slide with an antibody made against the antigen Yersinia, and
upon interaction, the complex is stained with fluorescence.
Transmission of Rickettsiae
Nymph Adult
Rarely to infect human Main human infection
Adherence.
Induced phagocytosis.
2) Phospholipase C:
Escape phagocytosis.
3) Recruitment of actin:
2) Typhus group
Epidemic Louse Borne Typhus Fever Endemic (Murine) Typhus
Headache, malaise, and myalgia. Maculopapular ( )حب صغير مفروشrash not
Rash begins on trunk not extremities. petechial; starts on trunk and spreads to
extremities.
2) Serologic tests:
Indirect fluorescent antibody (IFA):
Sensitive and specific.
Available in reference laboratories.
Immunofluorescence or immunoenzyme methods:
Examination of skin lesions.
Weil felix test (agglutination test):
Cross reacting antibodies.
Somatic antigens of non-motile Proteus.
Not reliable: low sensitivity and specificity.
3) Culture:
Cell cultures.
Difficult and hazardous.
Spotted Fever Disease Management & Treatment
Antibiotics:
Highly effective if given during the first week of illness.
Doxycycline for both children and adults.
Tetracyclines & Sulfonamides are contraindicated.
Affects on teeth color and late child growth.
Prevention:
Avoidance of tick contact.
Frequent tick removal in tick infested areas.
Vaccines are not licensed.
Trypanosomiasis
Hemoflagellates:
Site of diagnosis Intracellular Extracellular
The presence of flagella Absent Present
Classification:
There are main two species; Trypanosoma cruzi or brucei.
4) In the same way, a new insect feeds on the blood of the patient in the nocturnal
time to take the parasite and infect another human.
Note the life cycle inside the Gut of the insect:
Trypomastigotes epimastigotes trypomastigotes.
1) T.brucei:
T-cell independent:
2) T. Cruzi
Life cycle:
Mosquito salivary gland L3 Filariasis larvae
Human lymph L4 Male + Female
Human blood L1 Microfilaria
Mosquito thorax L2 Rhabditiform larvae
Life cycle events:
1) The insect injects from its salivary gland the infective form “Filariasis larvae”.
2) During 9-14 days it turns into a "L4".
After 6-12 months, the meeting takes place here and produces the male and
the female.
3) It produces a membrane and turns into a "microfilaria" during its passage into
the blood.
4) The insect picks up the infective form and enters the gut and then passes into
the thorax to become "Rhabditiform larvae".
5) It goes back to the salivary gland and belongs to type L3.
Clinical Aspects:
Acute manifestations:
Filarial fever.
Lymphadenitis, lymphedema, lymphangitis, epididymoorchitis.
Chronic manifestations:
Hydrocele,chyluria,elephantiasis.
Occult filariaisis:
Prevention:
Addition of DEC to salt.
Mosquito control.
Visceral Leishmaniasis:
Hemoflagellates.
Obligate intracellular parasites inside phagocytes.
Pathogenesis:
Symptoms are not visible; but within (3-12) months, it swells and becomes
reactivated, causing pigmentation.
Enlargement of liver, spleen, lymph nodes, bone marrow, small intestine, skin
pigmentation.
Double quotidian fever: twice/day, reappear at irregular intervals.
Diarrhea, malabsorption, weight loss.
Anemia, tachycardia, thrombocytopenia.
Leukocyte count < 𝟒𝟎𝟎𝟎/ 𝐦𝐦𝟑; agranulocytosis.
Elevated IgG, NOT PROTECTIVE.
Glomerulonephritis due immune complexes.
After the treatment, a macule is formed on the skin and then transformed into a
pigmented nodule to become its name; Post-Kala azar dermal leishmaniasis (PKDL).
Diagnosis:
Aspirate: bone marrow, liver, spleen, or lymph nodes.
Culture (N, N, N media).
Serological tests: DAT, ELISA, IFAT, rK39Ag test.
Virulence Factors:
Variable outer membrane proteins: due to recombination
between linear plasmids.
Use of manganese in place of iron in enzymes and electron
transport chain: Circumventing body’s natural defense
mechanisms -the lack of free iron in human tissues and fluids.
Use of SodA (Superoxide dismutase): metalloenzyme to
degrade ROS.
Not contain lipid A, but contains endotoxin similar lipoprotein.
OspA OspC
Bind to fibronectin (enhance
Depend on temperature, colonization) and serum factor H
pH), for binding to ticks (provide complement resistance,
midgut, low in human. protect against phagocytosis and
antibody).
Of course, OspA is little in humans, but in the case of arthralgia it is high.
Serum factor H is important in activating the alternative complement pathway, and
Borrelia inhibits it so that the classical pathway is activated.
Relapsing fever: Epidemiology
1) Endemic (tick-borne): Borrelia hermsii;
North America.
Reservoir: Rodents, rabbits, birds, lizards.
accidental host: Human
Infected tick Live years, trans-ovarial passage.
Diagnosis:
Microscopy: Giemsa or Wright staining of blood smears.
Spirochaetaceae shown in PBS.
Culture and serology: in reference laboratories.
Treatment:
Doxycycline or tetracycline (louse-borne) and alternatives; erythromycin
and ceftriaxone.
Risk of jarisch-Herxheimer; 24 hrs following treatment in high level of
bacteremia.
After treatment with antibiotics, Borrelia wall may rupture, accompanied by
endotoxin shock caused by the release of an endotoxin similar to
lipoprotein, and this results in several problems, including; Kidney failure,
heart failure, rash, headache and arthralgia.
Prevention:
Insecticide treatment.
Rodent control.
Improve hygiene.
Immunity:
Humoral; IgG develop after weeks or months after primary
infection.
Activation of classical complement.
No protective long lasting immunity.
Lyme disease: Manifestations, Diagnosis, Treatment & Prevention
Manifestation:
Bull's eye ring: annular lesion.
Erythema migrans.
Fluctuating arthritis: may become chronic.
Fever, fatigue, myalgia, headache, joint pains, and mild neck
stiffness, neurological or cardiac abnormalities.
Diagnosis:
Typical clinical findings.
Culture: need special media and experience.
PCR: B burgdorferi specific DNA.
EIA (screening Ab) confirmed by immunoblot (detect specific Ag): in later stages.
Immunoblot
1) There are special proteins secreted by Borrelia, we put them on the
electrophoresis device, and they separate and move from negative to positive,
forming a band.
2) We put the used gel on a cellulose filter paper.
3) We combine the filter paper with an antibody from the patient and another
marked antibody, where when the two antibodies interact together, the result
appears, and the first one alone (from the patient) does not give a result.
Treatment& Prevention:
Doxycycline and b-lactams (e.g amoxicillin, cefuroxime) and alternatives:
macrolides (azithromycin, clarithromycin).
No treatment for chronic cases.
Prevention: protective clothing, nymph removal, insect repellents, Prophylactic
doxycycline in endemic areas.
Francisella tularensis
Family: Francisellaceae.
Tiny, Gram negative coccobacilli.
Facultative anaerobe , encapsulated.
Cause: Tularemia (Rabbit fever).
Common in North America.
Morphologically similar Brucella.
Grow on Chocolate agar and Cysteine–glucose blood agar.
Zoonotic & arthropod born infection;
Skin bite, cut wound, inhalation, ingestion, Human-human (pneumonic
tolaremia).
Infective dose <100.
Vectors in animals Ticks and deer flies.
Reservoir: Ticks (transovarial)
Pathogenesis:
LPS not recognized by TLRs.
Escapes phagosome to macrophage cytoplasm.
Lesion at the site of infection, becomes ulcerated and early bacteremia may
occur.
Multiply within hepatocytes, kidney, alveolar epithelial cells and others.
It infects reticuloendothelial organs, forming granulomas.
Immunity:
T - cell–dependent: (either CD4+ or CD8+ cell) is the main resistant mechanism.
Natural acquired long lasting immunity is also developed (protective antibodies).
Life cycle:
A) During phagocytic ingestion of bacteria: the
bacteria interact with special components in the
phagocyte "early endocyst, late endocyst" that help it
to multiply inside the phagocyte and do not interact
with the lysosome.
B) It is released from the phagosome and replicated in
the cytosol.
There are two possible occurrences of infection:
1) Infecting more phagocytes after initial dead.
2) Invasion of various other cells and especially
(reticuloendothelial organs).
Life cycle:
Any of the three has the same mechanism, as well as affecting humans and
animals with the same mechanism.
The bartonella must pass by 2 niches; vascular endothelium and RBCs.
1) The insect injects Bartonella into the skin.
2) Macrophage or dendritic cells received it.
3) The macrophage transports the bartonella
to vascular endothelium then it released into
the peripheral circulation.
4) In the peripheral circulation the RBCs
invade and multiply within them.
5) After doubling, take one of two ways:
a) RBC lysis hemolysis.
b) Remain in the RBCs.
Transmission:
B. henselae B. quintana B. bacilliformis
Mainly zoonotic infection from Human body louse. Infected sand flies.
cat, but may directly from tick.
Epidemiology:
B. henselae B. quintana B. bacilliformis
Cat scratch Trench fever Oroya fever(Carrion’s
disease(CSD) disease)
Common in children Found only in Peru (endemic), Populations prone to
and adolescents, Ecuador, and Colombia, infestations with lice,
except for associated with a history of such as the homeless.
immunocompromised recent travel to these regions.
adults).
Trench fever:
It is often associated with poor hygiene areas and homeless.
It is normal to be in the form of a fever and ends within 4-5 days, but in some
people relapses (in 4-5 days) occur and may last for weeks.
Other complications: Bacteremia and endocarditis, headache, loin pain,
lumbago, knee and ankle pain, sever shin pain.
Oroya fever (Carrion’s disease):
Acute hemolytic anemia.
Nodular, highly vascular skin lesions called “verruga
peruana lesions”.
It is very similar to bacillary angiomatosis, but it is in the
lower extremities and not in the face.
Giemsa-stained blood smear showing parasitism of all
erythrocytes with B. bacilliformis.
Laboratory tests:
Serologic Demonstrate seroconversion.
testing IFA the most accurate for B henselae.
Microscopy Warthin-Starry silver stain.
The confirmatory stain for B henselae.
Molecular(PCR) Amplify ribosomal RNA gene fragments from tissue samples.
Culture Special agar medium to detect Bartonella quintana & B.
henselae.
Treatment &Prevention:
Reduce lymph node enlargement: Azithromycin or erythromycin
treat bacteremia in immunocompromised: Erythromycin or doxycycline
in Bartonella endocarditis: Valve replacement
Ehrlichia & Anapalsma
Anaplasmataceae family.
Obligate intracellular in WBCs.
Small gram negative.
Lack LPS and peptidoglycan.
Type Ehrlichia chaffeensis Anapalsma phagocytophilum
Vector Tick Tick
Reservoir Deer Rodent
Obligate in Monocytes Neutrophil
Associated diseases Human Monocytic Human Granulocytic
Ehrlichiosis (HME) Anaplasmosis (HGA)
Epidemiology:
HME southeastern and lower Midwestern United States
HGA Northern states, Asia and Europe.
Note that in the United States specifically, at the beginning we say that it is
Borrelia, and through the blood smear we can distinguish between it and
anaplasma;
Borrelia anaplasma
Soiralcheates Morula
Signs and Symptoms:
Manifestations may be mild.
Findings are clinically similar to Rocky Mountain spotted fever (RMSF), but
rashes are less commonly seen.
RMSF symptoms:
Rash in palms and soles.
Complications: DIC, thrombocytopenia, encephalitis, vascular collapse, renal,
heart failure.
May be life threatening: depending on the patient’s age and general health.
Flulike: fever, chills, nausea, muscle aches, and headache.
Leukopenia, thrombocytopenia.
Diarrhea, malaise.
Ehrlichiosis or anaplasmosis are considered in any case of unexplained acute
fever in patients exposed to ticks in endemic areas.
During the activation of the naïve B cells, monoclonal IgM (against the virus)
and also heterophile polyclonal IgM are secreted.
This heterophil IgM react with RBCs of some animals like; horse, sheep,
bovine and the agglutination is occur.
Trypanosoma and leishmania secrete heterophil IgM so nonspecific.
Manifestations of EBV:
Lymphoproliferative Syndrome:
Burkitt Lymphoma:
Treatment:
Supportive.
Corticosteroids: in Laryngeal obstruction as a result of lymphadenopathy.
Acyclovir: in Hairy leukoplakia in patients with AIDS.
Infections mononucleosis:
At the beginning of the disease there are no symptoms, but it develops in some
people (increases with AIDS) to mononucleosis disease.
Mononucleosis disease called also: glandular disease, kissing disease or mono.
Mononucleosis symptoms:
Fever.
Malaise.
Lymphadenopathy.
Sole throat.
Complication: ↑ liver enzymes, CNS, hepatosplenomegaly.
EBV-Immunity& Diagnosis:
1) Heterophile IgM antibodies are produced during an active EBV infection, not
specific.
The titer is not important, it is important to be positive.
2) Finding atypical lymphocytes and the heterophile antibody test are insensitive
for acute EBV infection, not specific.
3) IgM antibody to the EBV VCA antigen is useful in the acute phase.
VCA IgG and EBNA antibodies arise after the acute phase of illness.
4) Monospot test:
Serological test.
Take a blood from horse, sheep, or bovine and immerge
it with patient serum (IgM); if positive the agglutination
occur.
5) Downy cell test;
Infected T cells.
Large nucleus, large cytoplasm and irregular shape In
general.
Neither heterophile IgM nor downy cells determine EBV.
IgM increased in some cases:
In children; IgM before 4 years remain high and if EBV infect them the IgM titer
remain positive to almost 4 weeks.
In adult the IgM titer remain positive for more than 4 years then replaced by late
IgG.
In CBC of EBV:
Monocytosis.
Lymphocytosis.
↑ Bilirubin.
↑ Liver enzymes.
CMV
Beta herpesviridae.
Cause neonatal infection (one of TORCH infection).
TORCH stand for; Toxoplasma – rubella – CMV – herpes type VII.
With the above infections the IgM will be increase, increase risk of abortion
and congenital transmitted.
Cause cytopathic effects: Owl’s eye cells (nuclear inclusions) and cytomegaly.
In general, we do not search for the type of virus according to changing the
shape of the cell because it does not change it much, but in CMV virus, cell
shape changes and it is called (megalo) because it enlarge the cell.
The virus enters in the form of a nucleocapsid and in the cytoplasm leaves the
capsid and releases the tegument proteins, which include: PP71 and PP65.
PP65 PP71
contributes to viral Degradation of death domain-associated protein (Daxx)
replication and induce activation of the IE genes.
After the construction of the capsid and the DNA, they are assembled by the
nucleus and then go to the Golgi apparatus for the manufacture of envelope and
then exit from the cell.
Epidemiology:
Pathogenesis
Latency occur in the B cells, but may infect other type of the cells mainly
pluripotent stem cells (CD34) or monocytes.
Pluripotent stem cells (CD34) problem leukemia, pancytopenia.
The immune response may cause pneumonia.
Immunity:
In immunocompetent Primary infection occur, reactivation is subclinical.
In immunocompromised Primary infection and reactivation are symptomatic.
In allograft recipient Infected monocytes differentiate to macrophages once react
with activated T lymphocytes producing new CMV.
Manifestations:
Asymptomatic
Mononuleosis like syndrome in young adults.
Negative heterophile IgM.
Congenital defects (1st trimester):
Deafness ()الصمم, psychmotor mental retardation, hepatosplenomegaly,
jaundice, anemia, thrombocytopenia, DIC, pancytopenia, low birth weight,
microcephaly, and chorioretinitis ()التهاب المشيمية والشبكية.
In immunocompromised:
Interstitial pneumonia (following BM transplant), chorioretinitis, gastroenteritis,
neurologic disorders, and CMV retinitis (in AIDS patients).
Also EPV caused interstitial pneumonia.
Most study bind post-transplant organs infection with MCV.
CMV: Diagnosis
Culture: Detects cytopathic effect (owel’s eye).
PCR: Using plasma or leukocytes, high sensitive.
Biopsy: Demonstrate inclusions in tissues, best for CMV and
gastrointestinal CMV diagnosis.
Serogonversion: IgM, in non-immucompromised patients with primary infection.
Because in AIDS patients IgM already high.
CMV: Treatment and Prevention
Treatment:
Ganciclovir: inhibit viral replication
Immune globulin: for CMV pneumonia in bone marrow transplant recipients
Foscarnet: inhibits the CMV polymerase
Cidofovir: limited to ganciclovir resistant infections in immunosuppressed
patients.
Valganciclovir
Herpes virus 8
𝛾- Herpesviridae.
Known as KS-associated herpesvirus (KSHV).
Associated with primary effusion lymphoma (PEL) (100%) and multicentric
Castleman disease (MCD) with 50% of AIDS-related cases.
Endemic in Africa, some parts of Italy, Greece, Spain and Brazil.
The prevalence is more than 40% in adults.
Higher rates are seen in cohorts ( )مجموعةof men who have sex with men (MSM)
in the developed world and in association with HIV infection.
Transmission:
Manifestations:
Classic KS: Endemic KS:
strength Indolent Aggressive
Site of infection On lower extremities. on extremities, oral cavity
and torso
Epidemiology Mediterranean Ashkenazi jews. In central Africa.
Treatment:
Foscarnet and ganciclovir: inhibit lytic replication.
No treatment for latently infected cells or vaccine is available.
Parvovirus B19
Belong Parvoviridae family.
Naked virus.
The smallest virus “20nm”.
ssDNA linear.
Pathogenesis:
1) This virus invades immature nuclear red blood
cells.
2) The virus binds to the human P blood group
antigen.
3) There is a co receptor integrin to help it to invade
the nucleated red blood cell, which are not present
in the rest of the cells that express P antigen.
Life cycle:
1) Attachment and endocytosis.
2) Uncoating.
3) Host machine utilization:
Host DNA polymerase DNA replication
Host RNA polymerase DNA transcription
Clinical features:
Host Transmission
Infants-children Droplet infection (e.g., saliva)
Clinical features:
High fever.
May be with generalized convulsions (seizures).
Leukopenia.
Macular rash.