Exam 1 Flashcards

1
Q

Define hematology

A

The study of the structure and function of blood, blood forming organs, and their diseases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Microscopically identify elements of blood and blood forming tissues

A

Aqueous fraction - plasma
Acellular, water based
Plasma- uses anticoagulant
Serum- clotted plasma
Proteins in the plasma- albumin and globulins (ie. immunoglobulins, acute phase proteins, coagulation proteins, etc)
Electrolytes, nutrients, metabolic by-products, and signaling molecules are also found in plasma

Cellular fraction- RBC, leukocytes, and platelets

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is erythrocyte structure and function? What are species differences?

A

Fxn: oxygen delivery

In the millions per microliter

Structure- donut like with center divot (thinner in center); enucleated
Easiest to see in dig RBC
Goats have small RBC
Deer- sickle shaped RBC, artifact from blood reacting with oxygen
Alpaca and other camelids- elliptical
Non mammalians- nucleated RBC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are leukocyte functions and kinetics

A

Fxn: protection from exogenous (eg. infectious organisms) and endogenous (eg. cancer) harmful agents
In the thousands per microliter

Kinetics of neutrophils
Large number of neutrophils often needed in a short time
Storage pool necessary
Small animals- large pool
Large animals- small pool
Multiple factors stimulate production (IL-3, GM-CSF, G-CSF)
Most factors are produced by the cells of immune system
Glucocorticoids induce release of storage pool neutrophils → higher neutrophilia is seen in small animals as opposed to large animals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Leukocyte morphology- granulocytes vs mononuclear

A

Granulocytes or PMNs (polymorphonuclears)- cytoplasm has granules, nuclei are segmented and produce lots of different shapes; terminology reflects color when stained
Neutrophils- granules stain neutral
Eosinophils- granules stain orange
Basophils- granules stain blue-purple

Mononuclear- nuclei are not polymorphic, they are round
Lymphocytes
Monocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is platelet function and hemostasis

A

Fxn: blood coagulation
In the hundreds of thousands per microliter

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the appropriate technique of evaluation of blood and the blood forming tissues

A

Technique- blood must be maintain a liquid → use of an anticoagulant
Two components of coagulation- calcium ions (cofactors of many coagulation enzymes) and thrombin (a key protease)
EDTA (ethylene diamine tetra acetic acid)- used as an anticoagulant most often for a CBC, hematological purposes. Binds calcium to make it unavailable for clotting
Citrate- used as an anticoagulant most often for coagulation studies. It is also used to collect blood for transfusion purposes. Binds calcium to make it unavailable for clotting
Heparin- inhibits coagulation ny activating antithrombin, which inhibits the action of thrombin
Occasionally used for both hematology and blood chemistry analyses. Useful in small animals because unlike EDTA, plasma could be analyzed biochemically after hematological analysis

Hematologic evaluation
Hemogram or CBC
Consists of erythrogram, leukogram, thrombogram, and miscellaneous (plasma) sections
Bone marrow examination
Immune evaluation- Coomb’s test
Blood typing and cross match
Clotting studies
Flow cytometry
Relies on light (laser) interacting with cell
Generates shape and size of cell
Scattering of laser reflects internal complexity
After, stain the cells to determine type of cell
Or Blood smear and manually count (hemocytometer)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Define Hematopoiesis

A

production of all blood elements (ie. RBCs, platelets, and all leukocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Define myelopoiesis. What falls under this category

A

Myelopoiesis- production of non lymphoid bone marrow or bone marrow derived cells

Erythropoiesis- erythrocyte production
Granulopoiesis- neutrophil, eosinophil, and basophil production
Thrombopoiesis or megakaryocytopoiesis- platelet production

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Define lymphopoiesis

A

lymphocyte production

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is the lifespan of different blood cells

A

Neutrophils- 10 hour life span
Platelets- 10 day life span
RBCs- 100 day life span
In mammals, RBC life span is correlated with size
Lymphocytes- May live for many years

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Where are blood cells made in mammals? How does this change with age? How does this change when stressed?

A

Embryo- yolk sac (mesenchymal blood islands), liver and spleen
Fetus- liver, spleen, bone marrow (kidney, lymph nodes)
After birth- bone marrow
Young age- long and flat bones
Adults- flat bones and ends of long bones
Growing animals expand blood cell population. Adult maintain blood cell population
During stress, sites of fetal hematopoiesis can reactivate → extramedullary hematopoiesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Where are blood cells made comparatively?

A

mammals - bone marrow
Occurs extravascular
Once matured, go into sinus → blood vessel → go into circulation
Have megakaryocyte- big nucleated cells make platelets
Types of cells are intermixed together

Birds- bone marrow
Occurs extravascular but erythropoiesis and thrombopoiesis occurs inside of the sinuses
No megakaryocyte- have other cells but not morphologically different than their other cells → can’t see on slide
Types of cells are clustered together (ie. granulopoiesis in extravascular space, erythropoiesis within the sinusoid lumen)

Reptiles- bone marrow and spleen

Amphibians- spleen, kidney, and liver

Fish- kidney, spleen, and liver

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What factors stimulate blood cell production?

A

Humoral growth factors
Regulate proliferation and differentiation of bone marrow cells
Factors often act together in order to regulate production of a particular cell line
Some factors, may stimulate the production of a specific cell type, but inhibit the production of a different cell type
Liver- constitutive and inducible thrombopoietin
Kidney- inducible erythropoietin –> If there is renal failure, decreased erythropoietin and anemia is seen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is a hematopoietic microenvironment “niche”?

A

Hematopoiesis is regulated by a unique combination of structural, biochemical, nutritional, and cellular influences that develop or exist in bone marrow
Stromal cells, macrophages, endothelial cells, neurons, and the developing cells produce growth factors that influence that proliferation, commitment, differentiation, and maturation of developing cells
In addition, the matrix traps humoral growth factors and nutrients in the local area

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are proliferative, maturative, and storage bone marrow pools?

A

Conceptual pools

Progenitor compartment small cells- immature cell type; can’t tell which one is which

Precursor Compartment- recognizable cells (can see fate of cells)
Proliferative- cells are dividing and differentiating
Maturative pool- lost proliferative function, just maturing, differentiating
Storage- waiting in bone marrow, waiting to be pulled

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are changes as blood cells mature?

A

Cell size and nucleus size decrease (except megakaryocytic lineage)
Nucleus to cytoplasm ratio (N:C) decreases
Nucleoli disappear
Chromatin condenses
Basophilia of the cytoplasm decreases as RNA decreases
Specific cytoplasmic contents accumulate (ie. granules)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are features of erythropoietic precursor cells

A

Rubriblasts, prorubricyte, basophilic rubricyte- very blue cytoplasm, lots of rough ER to make protein
Polychromatic rubricyte- cytoplasm gets paler due to filling with hemoglobin (red color) while ribosomes still present (blue color)
Metarubricyte- gray/blue to red cytoplasm as even more hemoglobin, less RNA
Reticulocyte- no nucleus, pale blue-grey cytoplasm; residual RNA stains with new methylene blue
Same as polychromatic erythrocyte

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How are erythropoietic precursor cells characterized in the compartments in bone marrow

A

proliferative pool- rubriblasts, prorubricyte, basophilic rubricyte

maturative pool- polychromatophilic rubricyte, metarubricyte, reticulocyte

storage pool- no BM storage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are features of neutrophil precursor cells

A

Myeloblast- looks like rubriblasts, big round nucleus, euchromatin, nucleolus, small cytoplasm, blue cytoplasm
Proliferative pool
Progranulocyte- primary granules common
Myelocyte- primary granules go away and show up in this stage
Metamyelocyte
Band neutrophil
Mature neutrophil- nucleus fully segmented
Would be in maturation pool

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are some factors that stimulate neutrophilic production

A

G-CSF
Acts on progenitors, mitotic precursors
Increase number of neutrophils produced
Shortens production and maturation
Neutrophils ready sooner
Increases release of neutrophils from bone marrow
Gets more out of the bone marrow
Enhanced tissue emigration and functional capabilities
Used therapeutically

IL-5: stimulates eosinophil production

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What would glucocorticoids do in regards with neutrophils?

A

Glucocorticoids induce release of storage pool neutrophils → higher neutrophilia is seen in small animals as opposed to large animals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What are some facts about monocytopoiesis? Like about storage pools? Distinguishable stages?

A

CFU-GM: shared progenitor of neutrophils and monocytes
Cell lines diverge after CFU-GM
CFU-GM → monoblasts → promonocytes → monocytes
Monoblasts and promonocytes are difficult to distinguish from myeloblasts and promyelocytes
Monocytes do not stay in bone marrow: no storage pool
Early stages of monocytopoiesis cannot be confidently recognized morphologically
Monocytes become macrophages/histiocytes after leaving blood
Macrophages undergo many changes in tissue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What are platelets? How long do they live?

A

Platelets are cytoplasmic fragments from megakaryocytes
Highly complex cytoplasmic contents
Contain granules important in hemostasis
Can change shape
Live in circulation for about 6- 10 days

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

How is thrombopoiesis different than other blood cell productions and maturation?

A

Different because they get larger as nucleated precursors mature in the marrow
Mega- basophilic cytoplasm and 1-2 distinct nuclei
Endocytotic division

Pro- abundant RNA in cytoplasm → blue color cytoplasm
Continue endocytotic division
Cytoplasm increases in volume
Production of cytoplasmic contents occurs

Megakaryocytes (biggest) → break up into platelets

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

How is platelet production controlled?

A

Constitutive productive of TPO
Liver (and kidney)
Platelets bind TPO → inactivates
If normal platelet supports TPO binding → steady thrombopoiesis
Thrombocytopenia → more free TPO
Increased megakaryopoiesis
Not as many platelets to bind TPO
Increased TPO ⇒ increased platelet production in bone marrow
Thrombocytosis
Decreased megakaryopoiesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Does does platelet production differ among different species?

A

Birds, reptiles, amphibians, and fish have nucleated platelets
Thrombocytes- small cells with clear cytoplasm and small nuclei
Have few small granules in their cytoplasm
Produced in bone marrow vessel sinusoids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

How is transfer of thrombocytes from marrow to blood in birds?

A

In birds, erythrocytes and thrombocytes develop in marrow blind sac sinusoids
These cells do not have to move across the vessel walls to enter the blood
Thrombocytes simply leave the sinusoids and enter the blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What are common cellular elements of normal bone marrow?

A

Bone marrow is the primary hematopoietic organ in adults
2-3% of body weight

In young animals, the marrow of all bones is red due to hemoglobin
Active hematopoiesis

In adult animals, the mid-shaft marrow of long bones is yellow due to fat replacement of hematopoietic cells

Nutrient arteriole- surrounded by hematopoietic cells

Endothelial cells- interconnected cells lining the blood vessels

Granulocyte progeny

Stromal or reticular cell- produce hematopoietic short range regulatory molecules

Macrophage- engulf nuclei that extrude from erythroblasts when they turn into reticulocytes

Megakaryocyte- lies against veins and branches discharge platelets into vein
Send long pseudopodia into the blood
Cytoplasm of pseudopod fragments into platelets
Megakaryocytes shed platelets directly into the blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are the overarching functions of the immune system in health and disease

A

Protect the host from infections
Prevent invasion by pathogens
removal/inactivation of pathogens after infection
Protection from re-infection

Supports organ system health
Regulation of microbiota
“Housekeeping” functions such as removal of dead and dying cells
Tissue repair/remodeling

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What organs and cellular components are part of the immune system

A

Primary lymphoid tissues
Bone marrow- leukocyte development (hematopoiesis)
Fetal liver- development of leukocyte prior to birth
Thymus- generation of “T” lymphocytes

Secondary lymphoid tissues- localized responses
Spleen- filters blood
White pulp- filtering and removal of non-self components
Red pulp- filtering and removal of dysfunctional erythrocytes
Lymph nodes- filters lymphatics
Tonsils, peyer’s patches- filters antigens on surface of mucosal tissues of the respiratory (tonsils) and gastrointestinal (peyer’s patches) tract
Appendix- species specific functions

Tertiary lymphoid tissues- less developed than secondary but under right circumstances, can expand
Lymphocytic aggregates- develop after strong and/or chronic exposure to microbes (bacteria/viruses/parasites) or chronic inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are the fundamental differences between the innate and adaptive immune systems

A

Innate cell receptors- invariant in recognizing specific motifs or patterns and structures (Pathogen Associated Molecular Patterns and Danger Associated Molecular Patterns)
TLR4:LPS

Adaptive cells (lymphocytes)- active following binding to antigens
T and B lymphocytes recognize a specific stretch of primary amino acid sequences → three dimensional structure “conformation” of an antigen
These recognition motives on antigens are called epitopes
T cell- linear
B cell- any conformation
Difference is in the types of receptors they have

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Define CD markers

A

agreed upon naming system for proteins associated with cells, especially on cell surface
CD followed by a number referring to a unique structure/molecules on cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Define Antigen

A

antibody generation
Molecules that bind to specific receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Define antigen receptors

A

receptor on lymphocytes that binds to antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What are the principles of the immune cell activation

A

Tissue resident innate immune cells will secrete chemokines and cytokines that activate endothelial cells to express selectin ligands
Leukocyte express selectins and activation changes expression of chemokine receptors. Endothelial cells express selectin ligands after injury. Leukocytes start slowing down “rolling”
Slowing down of cells allows them to bind to selectin ligands and chemokines, which will lead to activation of integrins
Activated integrins bind to endothelial cells, which causes firm adhesion of the cell
The cell will transmigrate from blood into tissues following a chemokine gradient

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

How are leukocyte migration pattern connected to their function and activation state

A

In health, they continuously move through arterial and venous blood and in and out of secondary organs → into non-lymphoid tissues and can return into circulation
If they get a signal (chemokine, cytokine), leukocytes are instructed to leave the blood and either get sequestered in secondary lymphoid tissues and/or enter a tissue/organ
Migrated to tissue- activated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Describe the general structure of hemoglobin and how binding to oxygen is regulated

A

Four peptide chains together → 2 alpha and 2 beta chains
Each chain has a heme → each each has iron
Heme is a porphyrin ring, with iron in the middle

Iron has two states
Ferrous- 2+ “for us” → used for breathing
Ferric- 3+ “icky” → oxidized and can’t be used for oxygenation

Regulation→ affinity for oxygen is adjustable to tissue environment (Bohr environment)

Lower pH and lower O2 content (eg. in actively metabolizing tissues) facilitates unloading of oxygen (lower affinity)
Decreased pH
Increased 2-3 DPG
Increased CO2
Increased temperature
Shift to the right

Higher pH and higher O2 content (eg. in the lungs) facilitates loading of oxygen (higher affinity)
Increased pH
Decreased 2-3 DPG
Decreased CO2
Decreased temperature
Shift to the left

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Name the main metabolic pathways in the red cell and their primary function

A

Methemoglobin Reductase Pathway- ensuring Fe stays in ferrous state and not ferric state
Ferric iron = methemoglobin → lose all oxygen carrying capacity
NADH takes the charge from ferric → becomes NAD+ and ferrous
Always on and can be overwhelmed

Rapoport-Luebering pathway- 2,3 DPG synthesis → regulates hemoglobin oxygen affinity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Define the major steps in hemoglobin synthesis; give an example of a disease that interferes with synthesis

A

Highly complex, involving organic compounds
Some steps happen in mitochondria and some in cytoplasm

Abnormalities in heme synthesis due to
Porphyrias (inherited enzymes defects in porphyrin synthesis)- very rare; lowers RBC and oxygen carrying capacity
Dyserythropoiesis (acquired defects)- due to toxin or tumor

Heme synthetase and Fe2+ - sticking heme onto RBC
Dyserythropoiesis, specifically lead toxicity leads to this shutting down

ALA dehydratase- cleaves off water from components
Lead toxicity deactivates this

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Outline the basic steps and key molecules involved in iron absorption, utilization, and recycling

A

Iron absorption through diet and intestinal absorption
Iron and heme absorbed through enterocyte → broken down into ferrous iron → have three final outcomes
Ferritin: iron storage protein in macrophages (short term)
Hemosiderin not really accessible but also in macrophages
Ferroportin: iron “portal” protein, regulated by hepcidin (hormone)
Master regulator of iron across body
Increased hepcidin ⇒ decreased ferroportin ⇒ less iron transported in
Decreased hepcidin ⇒ increased ferroportin ⇒ more iron transported in
Synthesized in the liver
Adjusts to changes in body iron stores
Adjusts to changes in erythrocytes (hemoglobin synthesis)
Released in inflammation
Blocks release of iron absorption in the intestine and blocks release of iron from macrophages to developing erythroid cells
Transferrin: iron transport protein in serum
Can bring it to a macrophage to be stored

Utilization in erythropoiesis
Iron travels in plasma and binds to bone marrow macrophage → binds to transferrin receptor → internalizes Fe
Can be stored in ferritin (short term) and hemosiderin (long term) storage
Can be transferred through macrophage and ferroportin protein (regulated by hepcidin) to erythroid precursor cells

Iron recycling from senescent red cells
Circulating RBCs in peripheral blood → erythrophagocytosis of senescent (old) RBCs → in macrophage → heme is broken down → iron is taken from it and can go in the three pathways

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is “RBC mass” and how is it measured

A

HCT= hematocrit (%)
Percent blood occupied by RBC → extrapolated
Spun microhematocrit (PCV)
Calculated HCT= RBC count x MCV (size)
Most representative of mass

RBC= red cell count (number of RBCs/microliter)
Not much emphasis as it could be misleading → having tiny or big RBC can lead to inaccurate count

HGB= hemoglobin concentration (g/dl)
Used more in human medicine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Describe the main components of the red cell membrane and their functional purpose

A

RBCs change shape as travel capillaries → RBC membrane must be deformable
ATP provides energy for membrane contractile proteins → change the shape RBC and return its normal shape when RBC reenters larger vessels

Cytoskeleton (structural proteins)- in cell and anchored to the membrane
Help determine and maintain RBC shape
Viscoelastic properties
Delimit deformability
Cross linked or damage = loses elasticity

Lipid bilayer- provides anchor point
Permeability barrier
In equilibrium with plasma lipids → plasma proteins can alter RBC membrane → alters function and flexibility
Noncompressible
Outer membrane- a lot of CHO → makes it tough so it can take damage
Too tough or too weak → lysis
Inner membrane- ion channels → move ions so that internal solutes = external solutes for osmotic pressure

Membrane components (horizontal and vertical)
Spectrin- spider web; really strong; can move all around; allows them to bend
Actin- facilitates bending (flexing) → proactive movement
Vertical- points of anchor help vertical interactions without lysing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Explain the major causes and implications of abnormalities involving the red cell membranes

A

Embden-meyerhof pathway
Relies on ATP production for membrane shape, cation pump, and ionic gradient
Maintains cell function and size
Maintain osmotic balances
Abnormalities- can lead to water going into cell → lysis or water going out of cell

Membrane abnormalities
Structural abnormalities (in lipids or proteins)
Metabolic abnormalities (often involve ATP, Ca2+)
These can lead to
Decreased deformability → lysis
Shape changes (poikilocytosis)- more prone to damage or not bending
Premature destruction (hemolysis)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is a poikilocyte?

A

abnormally shaped red cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What is an echinocytes (crenation)? What is the mechanism of formation?

A

has pointy edges, smaller, evenly spaced and shaped

Normal or artifact: Drying artifact in smear preparation; prolonged storage in EDTA
Pathologic: electrolyte abnormalities, uremia, rattlesnake envenomation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What is an acanthocyte? What is the mechanism of formation?

A

big, rounded projections, larger and more irregular compared to echinocytes

Normal: pigs, calves, rabbits
Pathologic: hepatic disease, lipid abnormalities, red cell fragmentation, neoplasia → incorporations of too little or too much CHO into the outer leaflet of the lipid bilayer causes irregular membrane protrusions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What are target cells? What is the mechanism of formation?

A

lose biconcase nature

Normal or artifact: drying artifact in smear preparation
Pathologic: anemia (nonspecific), iron deficiency, hepatobiliary disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What are schistocytes? What is the mechanism of formation?

A

tiny fragments of RBC torn apart and membrane reanneals

Normal: none
Pathologic: red cell fragmentation → seen in small or large clots → fibrin is really tough → results in cleavage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are keratocytes? What is the mechanism of formation?

A

Normal: none
Pathologic: red cell fragmentation → membrane blisters and it goes outwards
Common in bad trauma → results in a lot of blood clots

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are spherocytes? What is the mechanism of formation?

A

very rounded than normal; lose biconcave nature

Normal: none (rabbits may have a few normally)
Pathologic: immune-mediated hemolytic anemia (when spherocytes are the only poikilocyte)
Agglutination- crosslinking of antibodies on RBC surface and causes them to clump together → with spherocytes, it is a hallmark of IMHA
Pathologic: RBC fragmentation (when few spherocytes and schistocytes and acanthocytes, etc) → part of RBC membrane is removed by macrophage and remaining membrane reseals around the cytosol

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are Heinz bodies? What is the mechanism of formation?

A

Oxidation of globin chains, which clump and bind to the inner red cell membrane, protruding from the surface
Cause- oxidant drugs, plants, chemicals
Feline HGb is oxidant-sensitive
High number of SH groups
Up to 5% of RBCs may contain Heinz bodies (more with some fish diets)
Staining- hard to detect with eyes but when stained with methylene blue → you can see

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What are eccentrocytes? What is the mechanism of formation?

A

less common than Heinz

Internal structure membrane oxidizes → stick together and squishes the cell → hemoglobin is pushed onto one sude
Normal: none
Pathologic: oxidative damage (+// Heinz bodies, eg. onion induced hemolysis)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Diagram the pathway of red cell and hemoglobin degradation

A

Metabolic functions fails, ATP is depleted
Reducing power fails, gradual oxidation of hemoglobin
“Senescence” antibodies bind to altered membrane proteins
Phagocytosed by splenic macrophages
RBC enters macrophage → globin is broken down into AAs and heme is broken down into iron and other elements → bilirubin (chemically inert)→ bili put on mac surface → albumin takes the bili → takes to hepatocyte (liver) → liver sticks sugar on it → soluble in water and gets defecated in health
In disease, hyperbili → bilirubinemia → get rid of it through GI or kidney → bili in urine (biliuria)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Describe where plasma proteins are synthesized

A

Albumin- maintains oncotic pressure; also a carrier protein
Synthesized by hepatocytes (liver)
Highest concentration of any single protein in plasma
~65 kDa (small)

Globulins

Immunoglobulins (gamma)- antibodies produced by adaptive immune cells
Synthesized by plasma cells
Account for most of the globulins in plasma
150-970 kDa (big)

Other globulins (alpha, beta)- roles in innate inflammatory response
Synthesized by hepatocytes (liver)
Acute phase proteins; transport proteins; complement; clotting proteins; lipoproteins
Hepcidin falls under this category
Variable kDa (small)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Explain the difference between plasma and serum. How do we measure proteins in them?

A

Plasma- includes all proteins, including fibrinogen and clotting factors
Purple top tube (CBC)- EDTA anticoagulant
Proteins levels always higher compared to serum
Plasma protein, plasma fibrinogen, and protein:fibrinogen ratio measured
Refractometry- refractive index correlates with solute concentration
Accurate to +/- 0.1 g/dl
High concentrations of glucose, sodium, or urea can falsely increase TPP
Hemolysis and lipemia can interfere with reading

Serum- fibrinogen and clotting factors are absent, consumed in the clot
Red top tube (chem panel)- no anticoagulant
Total protein- biuret method
Highly specific and sensitive
Albumin- bromocresol green method
Species differences in dye binding (doesn’t work in birds)
Bilirubin and some drugs interfere with binding
globulin= (total protein - albumin)
a/g ratio calculated to detect disproportionate changes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Describe the main pathophysiologic mechanisms for hyperalbuminemia

A

dehydration (relative)
Globulins also increase proportionately
The liver never synthesizes too much albumin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Describe the main pathophysiologic mechanisms for hyperglobulinemia

A

Increased production of gamma globulins (hypergammaglobulinemia)

Chronic inflammation (commonly seen)
Increased immunoglobulin (antibody) production by plasma cells due to chronic antigenic stimulation
Albumin WRR
Polyclonal gammopathy → broad based gamma peak

Lymphoid neoplasia
Abnormal immunoglobulin (paraprotein) produced by neoplastic plasma cells or B lymphocytes
Low albumin occasionally
Monoclonal gammopathy → narrow based gamma peak

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Describe the main pathophysiologic mechanisms for atypical gammopathies

A

Occasionally, infections can cause a monoclonal spike (+/- polyclonal)
Canine ehrlichiosis - most common
Leishmaniasis
Rarely other lymphoplasmacytic inflammatory lesions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Describe the main pathophysiologic mechanisms for hyperproteinemia

A

can result from increased albumin, increased globulins, or both
Hyperalbuminemia with normal or increased globulins → dehydration
Hyperglobulinemia with normal or decreased albumin → chronic inflammatory disease* or lymphoid neoplasia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Describe the main pathophysiologic mechanisms for increased production of alpha and beta globulins

A

Acute phase response
Cytokine-mediated production of “Acute phase proteins” - mostly in the liver
Most of these proteins are in very low concentrations in plasma, so increases do not usually affect total protein or globulin concentrations
Also increases fibrinogen and decreases albumin (minor)

Hyperfibrinogenemia
Fibrinogen is the most abundant acute phase protein
Increases in fibrinogen can increase the total plasma protein concentration
Acute (and chronic) inflammation
Large animals
Chronic inflammation
Small animals, birds
Prot:Fib ratio
Proportionate increase in dehydration (more than or equal to 15)
Disproportionate increase in fibrinogen is inflammation (ratio less than 15)
Remember to convert mg → g (divide by 1000)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Describe the main pathophysiologic mechanisms for hypoalbuminemia

A

Decreased synthesis
Hepatic insufficiency
Acute phase response
Compensatory response to marked hyperglobulinemia

Increased loss
Renal loss (glomerular) → urine
Gastrointestinal loss (diarrhea, malabsorption)
Blood loss, exudates, vasculitis

Decreased intake and increased utilization/catabolism (malnutrition/cachexia)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Describe the main pathophysiologic mechanisms for hypoglobulinemia

A

Decreased synthesis
Immunodeficiencies (hypogammaglobulinemia)
Hepatic insufficiency (decreased alpha, beta globulins)

Increased loss
Renal loss (small globulins are lost in severe tubular disease)
Gastrointestinal loss (diarrhea, malabsorption)
Blood loss, exudates

Decreased intake (failure of passive transfer)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Describe the main pathophysiologic mechanisms for hypoproteinemia

A

Can result from hypoalbuminemia, hypoglobulinemia, or both
Panhypoproteinemia (hypoalbuminemia + hypoglobulinemia) → liver failure or loss
Hypoalbuminemia with normal or increased globulins → primarily albumin production issue
Hypoglobulinemia with normal or increased albumin → primarily globulin production issue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What is neutrophil function?

A

primary first line defense versus bacterial infections- acute inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What is neutrophil structure?

A

Mature neutrophils have multiple nuclear lobes separated by constrictions (filaments)
Contain cytoplasmic lysosomal granules that take up a little stain (with routine stains)

Primary or azurophilic granules
Larger and more electron dense than secondary granules
Stain red/purple with Romanowsky stains but don’t stain and aren’t visible after the promyelocyte stage in many species
Contents include proteases, acid hydrolases, peroxidase, lysozyme, microbicidal cationic proteins, esterases, glycosaminoglycans
The microbicidal properties of neutrophils are contained primarily in the lysosomal granules
Degranulation results in release of enzymes and proteins
Oxygen dependent or independent

Secondary or specific granules
Usually not visible (in neutrophils) with Romanowsky stains
Contents include lysozyme, lactoferrin, collagenase, plasminogen activator, phospholipase A

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What is eosinophil and basophil function?

A

important in protection against helminth infections and participate in allergic reactions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What is monocyte function?

A

become macrophages (in tissue); key players in phagocytosis
Antigen processing and presentation
Production of inflammatory mediators and cytokines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is lymphocyte function?

A

antibody and cytokine production; mediators in destruction of microorganisms and tumor cells
distinguish between self and non self
Responsible for memory
Humoral and CMI

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Are there species differences when it comes to blood cells?

A

In some animals, heterophils instead of neutrophils
Heterophils in rabbits, guinea pigs, birds, and reptiles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What are leukocytes

A

nucleated cell that travels through the blood to get to the tissues, where it functions (literally, means “white cell”, based on its unstained appearance)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What is a leukon

A

all the leukocytes in the body including 1) marrow precursors, 2) leukocytes in the blood, 3) leukocytes and leukocyte derived cells in the tissues

73
Q

What is a leukogram?

A

blood test evaluating leukocytes, usually part of the CBC
All nucleated cells counted, including nRBC’s

74
Q

What is leukocytosis

A

increase in leukocyte numbers above the reference limit (neutrophilia or heterophilia is the usual case)
Because neutrophils are the most common leukocyte
Heterophils in rabbits, guinea pigs, birds, and reptiles

75
Q

What is leukopenia

A

decrease in leukocyte numbers below the reference limit (neutropenia is the usual case)

76
Q

What is panleukopenia?

A

decrease in all leukocyte types in the blood below their respective reference limits

77
Q

What is pancytopenia?

A

decrease in all blood cell types (leukocytes, rbc’s, platelets) in the blood below their respective reference limits

78
Q

Which leukocytes are considered granulocytic leukocytes (granulocytes)

A

neutrophils (heterophils), eosinophils, basophils

79
Q

Which leukocytes are considered mononuclear leukocytes?

A

lymphocytes, monocytes

80
Q

What are the mechanisms and kinetics impacting circulating leukocyte numbers?

A

Infections specific to leukocytes
Neoplasia involving leukocytes
Inherited conditions specifically affecting leukocytes
Multisystemic inherited diseases (storage diseases), including leukocytes (leukocytes are useful marker for some of these diseases

81
Q

What are the major causes and players in the regulation of granulopoiesis?

A

The major players are IL-3, GM-CSF, and G-CSF (primary specific regulator)
IL-3 and GM-CSF- potent stimulator of all leukocytes
Act directly on the bone marrow precursors ro
Increase stem cell commitment and cellular proliferation
Promote neutrophil differentiation and maturation
Result in differentiated cellular function

Inflammation results in release of cytokines by activated T cells, macrophages, endothelium, and other cells
In concert with other inflammatory mediators (IL-1, TNF), also stimulate marrow release into blood and emigration of neutrophils into tissues

The (essential) net result is to increase the numbers of functioning neutrophils at the site of injury

82
Q

Define anemia and expected bone marrow response to hypoxia, including species differences in the regenerative response

A

Anemia- decreased red cell mass
Decreased RBC mass → hypoxia (lower blood oxygen tension in kidney) → erythropoietin production → stimulation of erythropoiesis (increased red cell production) → release of reticulocytes and replaces RBC mass (regenerative response)

Regeneration depends on
Severity of anemia (hypoxic)
A lower HCT (more hypoxia) elicits increased response
Duration of the anemia
Ability of the bone marrow to upregulate erythropoiesis

83
Q

What are the major types of anemia, their pathophysiology, and how to distinguish between them?

A

Anemia (general)
Low HCT, low Hgb, low RBCC

Regenerative- high MCV and low or WRI MCHC
Regeneration evidence- high anisocytosis (and RDW), high polychromasia (and high reticulocyte count), high or normal MCV, low MCHC, high nucleated RBCs, and +/- basophilic stippling

Most non-regenerative- WRI MCV and WRI MCHC
Mechanisms- Decreased erythropoiesis and ineffective erythropoiesis
Causes- extramedullary cause (system disease process→ could be anything and everything since the erythropoiesis is so complicated) and medullary cause (bone marrow disease, pure red cell aplasia, immune mediated anemia → slowing down production or ineffective)
Most common cause is chronic inflammation
Decreases EPO, decreases RBC lifespan, hepcidin release, iron sequestration, and decreased erythropoiesis
Medullary causes are less common
Bone marrow destruction/altered hematopoietic precursors and niche- often affects myeloid, erythroid, and megakaryocytic lineages
Concurrent cytopenias → often severe
CBC- schistocyte, keratocyte, dacrocytes, elliptocytes; atypical nRBCs, myeloid cells, and lymphoid cells
Bone marrow- highly variable findings
Features- absence of regeneration (minimal polychromasia) and RBC shapes (usually none)

Iron deficiency: hypochromasia, target cells, schistocytes, keratocytes
Bone marrow disorder: Elliptocytes, dacrocytes, schistocytes, keratocytes

Hemolysis- high MCHC
Seen with high bilirubin, bilirubinuria, hemoglobinemia, and hemoglobinuria
Three different causes
Trauma- external wounds and bruising
Alimentary- GI blood loss
Acute will be regenerative
Chronic will cause iron deficiency → degenerative anemia
Hemostatic abnormality- issues with platelets and coagulation factors
Types of hemolysis
Immune-mediated
Antibodies bind and coat RBC → splenic macrophage phagocytosis the RBC (extravascular) or complement cascade punches hole into it to lyse (intravascular) or crosslink (agglutination)
Seen in spherocytes and agglutination
Could be microorganisms in RBC → triggers immune response → target RBCs and they get damaged
Oxidative
Heavy oxidative materials cause RBCs to become Heinz bodies → harder to bend and they lyse when they bend
Become ghost cells → after lyse, just see faint remnants
Eccentrocytes also seen
Exogenous
Heavy metals*
Acetaminophen (tylenol)
Skunk musk (sulfur intake)
Allium family plants (onions)
Endogenous- build up of internal things
Ketoacids
Uremic acids
Hyperthyroidism
Mechanical lysis
Keratocytes and schistocytes
Disseminated intravascular coagulation, hemangiosarcoma, vasculitis, and thrombi
Iron deficiency- low MCV and WRI or low MCHC
Fe needed for Fe synthesis
Cellular Hgb guides RBC division
Causes- Chronic hemorrhage*, dietary deficiency (less Fe and Cu)
Morphology- hypochromasia, schistocytes, keratocytes, thrombocytosis
Over time, cells become more microcytic and hypochromic
Extra cell divisions and less Fe

84
Q

How to discern regenerative anemia

A

high MCV and low or WRI MCHC
Regeneration evidence- high anisocytosis (and RDW), high polychromasia (and high reticulocyte count), high or normal MCV, low MCHC, high nucleated RBCs, and +/- basophilic stippling

85
Q

How to discern non regenerative anemia

A

WRI MCV and WRI MCHC

Mechanisms- Decreased erythropoiesis and ineffective erythropoiesis

Causes- extramedullary cause (system disease process→ could be anything and everything since the erythropoiesis is so complicated) and medullary cause (bone marrow disease, pure red cell aplasia, immune mediated anemia → slowing down production or ineffective)
Most common cause is chronic inflammation
Decreases EPO, decreases RBC lifespan, hepcidin release, iron sequestration, and decreased erythropoiesis
Medullary causes are less common
Bone marrow destruction/altered hematopoietic precursors and niche- often affects myeloid, erythroid, and megakaryocytic lineages
Concurrent cytopenias → often severe
CBC- schistocyte, keratocyte, dacrocytes, elliptocytes; atypical nRBCs, myeloid cells, and lymphoid cells
Bone marrow- highly variable findings

Features- absence of regeneration (minimal polychromasia) and RBC shapes (usually none)
Iron deficiency: hypochromasia, target cells, schistocytes, keratocytes
Bone marrow disorder: Elliptocytes, dacrocytes, schistocytes, keratocytes

86
Q

How to discern hemolysis

A

high MCHC
Seen with high bilirubin, bilirubinuria, hemoglobinemia, and hemoglobinuria
Three different causes
Trauma- external wounds and bruising
Alimentary- GI blood loss
Acute will be regenerative
Chronic will cause iron deficiency → degenerative anemia
Hemostatic abnormality- issues with platelets and coagulation factors

87
Q

What are the different types of hemolysis

A

Immune-mediated
Antibodies bind and coat RBC → splenic macrophage phagocytosis the RBC (extravascular) or complement cascade punches hole into it to lyse (intravascular) or crosslink (agglutination)
Seen in spherocytes and agglutination
Could be microorganisms in RBC → triggers immune response → target RBCs and they get damaged

Oxidative
Heavy oxidative materials cause RBCs to become Heinz bodies → harder to bend and they lyse when they bend
Become ghost cells → after lyse, just see faint remnants
Eccentrocytes also seen
Exogenous
Heavy metals*
Acetaminophen (tylenol)
Skunk musk (sulfur intake)
Allium family plants (onions)
Endogenous- build up of internal things
Ketoacids
Uremic acids
Hyperthyroidism

Mechanical lysis
Keratocytes and schistocytes
Disseminated intravascular coagulation, hemangiosarcoma, vasculitis, and thrombi

88
Q

How to discern iron deficiency

A

low MCV and WRI or low MCHC
Fe needed for Fe synthesis
Cellular Hgb guides RBC division
Causes- Chronic hemorrhage*, dietary deficiency (less Fe and Cu)
Morphology- hypochromasia, schistocytes, keratocytes, thrombocytosis
Over time, cells become more microcytic and hypochromic
Extra cell divisions and less Fe

89
Q

What are the major mechanisms of polycythemia? How do you distinguish them?

A

Relative (common) of erythrocytosis
Dehydration: HCT + plasma proteins increase
Splenic contraction
EPI induced, mainly horses and cats
HCT and platelets increase

Absolute (rare)
Primary (RBC neoplasia = polycythemia vera)
Secondary (EPO secretion due to chronic hypoxia or by rare tumors)

90
Q

Recognize cells and soluble molecules of the immune system. What is the cell’s identity as part of the innate or adaptive arm of the immune system?

A

Innate
Macrophage
Neutrophil
Dendritic cell- link innate and adaptive
Eosinophil
Basophil
Natural killer

Adaptive
Lymphocytes

Chemokines, cytokines- pro + anti inflammatory; chemoattractant or chemorepulsion

91
Q

What are the principles of innate immune recognition of antigens?

A

Breach of barrier
Recognition by sentinels
Recruitment of reinforcement
Pathogen elimination and resolution

92
Q

In innate immune recognition, what consists of the breach of barrier step?

A

Mechanical barrier- epithelia form specialized barriers; form tight junctions; differ based on the physiological requirements of tissue

Chemical barrier- antimicrobial substances (lysozyme, deferins, cathelicidins,…)
kills/inhibits microbes

Microbial barrier- resident flora prevents colonization of pathogenic bacteria

93
Q

In innate immune recognition, what consists of the recognition by sentinels step?

A

Dendritic cells and macrophages act as sentinels at epithelial barriers
Mac- phagocytic cell that consumes foreign pathogens and cancer cells. Stimulates response of other immune cells
Migrates from blood vessels into tissues
Origin- in embryonic development, made in yolk sac and fetal liver → migrate to tissue and regenerate; in adults, made in bone marrow, generate monocyte → in bloodstream and becomes macrophage in tissue
Dendritic cell- presents antigens on its surface, thereby triggering adaptive immunity
Present in epithelial tissue, including skin, lung, and tissues of the digestive tract. Migrates to lymph nodes upon activation

Macrophage surveillance (steady state vs. injury)
In healthy tissue, macrophages take up and clear apoptotic cells etc. without recruiting other cells by using scavenger receptors
Skin and GI: macrophages are located behind epithelial barrier
Lung (lower airways): No great physical barrier present, alveolar macrophages is in front of epithelium/in lumen of alveoli
Liver- kupffer cells and CNS- microglia
Survey for damage and are first responders following an insult

Sensory organ of the cell is the receptor → antigen recognition
Innate- one cell has many receptor types and many specificities (not specific)
Recognition of conserved epitope (ubiquitous) → present on different pathogen
Using pattern recognition receptors (PRRs)
Can mass produce cells since all of them have similar antigen recognition → identical genomes

Exogenous ligands → PAMPS- repetitive structures that are found across classes of pathogens (bacteria, viruses, fungi, etc)
Signal “stranger” and are recognized by pattern recognition receptors (PRRs)
Toll like receptor- phylogenetically conserved PRR family
Surveil compartments- extracellular, cytosolic, endosome
Triggered when encountering molecules that either don’t exist in the host (eg. dsRNA) or are not normally present in a given compartment (eg. ssRNA)
Endogenous ligands → DAMPS- released by cellular trauma (physical damage- cuts, tears, etc or damage by heat/cold)
Release of cellular materials into the extracellular environment
Endogenous molecule but in the wrong compartment

Altered self→ natural killer cells
Major function- cytotoxicity = destruction of target cells
Bind Major Histocompatibility Complexes (MHC) on the surface of the cells
NK receptors are either activating or inhibitory
Lack of killing requires an inhibitory signal from the target cell
Killing occurs by pore formation (perforin) and release of cytotoxic granules (perforins)
Steady state
Balance between inhibitory and activating signal from target cell
NO target cell destruction
Cancer or viral infection
Downregulation of inhibitory signal on target cell (MHC I)
Activating signal dominates
Cell lysis

94
Q

In innate immune recognition, what consists of the recruitment of reinforcement step?

A

TLR engagement leads to activation of transcriptional regulator NF-kB
Signal transduction is produced → causes cascade of signaling molecules and changes in gene expression
One of the responses is release of cytokines and chemokines
Pro-inflmmatory- released by macrophages upon activation via PRR; stimulate inflammation
Tumor necrosis factor (TNF alpha)
Interleukin 1 (IL-1)
Interleukin 6 (IL-6)
anti-inflammatory cytokines: released by engagement of scavenger receptors during healing phase of response
Transforming growth factor beta (TGF beta)
Interleukin (IL-10)

Acute phase response in liver
APPs are a class of proteins whose plasma concentrations
Increase positive acute phase proteins or
Decrease negative acute phase proteins
In response to inflammation
Roles- opsonization, complement activation, chemotaxis, and trapping of microbes
Extravasation of leukocytes- vasodilation and induction of adhesion molecules → rolling → adhesion → extravasation/diapedesis → migration
Neutrophils: within 6 hour of inflammatory response; chemokine CXCL8
Macrophage: more slowly; chemokine CCL2

95
Q

In innate immune recognition, what consists of the pathogen elimination and resolution step?

A

Sentinel cells in different tissues (liver and CNS)
Antigen recognition
Signal transduction and chemotaxis to tissue
Kill all pathogens

By phagocytosis
Microbe is bound by receptor on surface of phagocytosis
Internalization
Fusion with lysosome → phagolysosome
Formation of oxide radicals → killing

By NETosis- neutrophil Extracellular Traps
Regulated form of neutrophil cell death
Nuclear chromatin is released into extracellular space creating neutrophil extracellular traps (NETs)
NETs capture microbes and facilitate phagocytosis by macrophages and neutrophils

By complement system
Enhances the ability of antibodies and phagocytic cells to clear microbes and damaged cells from an organism, promote inflammation, and attack the pathogen’s cell membrane
Origin- proteins synthesized in liver, present in blood as inactive precursors, and is 10% of the globulin fraction of blood serum
C3a- inflammation
C3b- opsonization and phagocytosis
Increases vascular permeability
Causes gaps in the endothelium for components of blood to go inside
C5a- inflammation

96
Q

What is neutrophil function?

A

Phagocytosis and microbial action
Done through adherence and emigration through vessel wall- involves rolling, sticking (adhesion), and extravasation via selectins, integrins, chemokines, and ICAMS
Chemotaxis- C5a, C567, FActor XIIa, LTB4, IL-8 (specific for neutrophils (CXCL8), PAF, bacterial products
Ingestion, degranulation, and bacterial killing

Bacterial killing
Oxygen independent (microbicidal cationic proteins - defensins, cathepsins, BPI, etc)
Oxygen dependent
Respiratory “burst” and activation of NADPH oxidase
Hydrogen peroxide (H2O2)
Superoxide anion (O2-)
Hydroxyl radical (OH)
Singlet oxygen (O2)
These ROS denature proteins, oxidize lipids ie. bactericidal
H2O2 complexes with MPO (myeloperoxidase) to form a VERY potent bactericidal system MPO-H2O2-Halide complex that results in production of HOCL, a powerful toxic radical
There are protective mechanisms to “detoxify” reactive species (SOD, catalase, GPx/GR system, NADPH)

97
Q

How does neutrophil kinetics work?

A

Compartment or “pools”
Progenitor pool (stem cells, committed stem cells)
Proliferative pool (~20% of precursors, undergo mitosis, 4-5 mitoses, some attrition at myelocyte stage, transit time (tt), ~2.5 days)
Maturation-storage pool (80% of precursors, don’t undergo mitosis, tt~2.5 days, size of storage pool varies with spp. → dogs have biggest storage pool, oldest cells enter the blood first)

Sequential unidirectional movement through pools
Size of pools and rate of traffic between them determines both number and degree of maturity of neutrophils in blood

Marginal pool (MP)- neutrophils accumulate near the walls of small vessels (capillaries, post capillary venules)- attach, roll, detach intermittently before migrate into tissues
Random exchange between MP and CP
Marginal pool cells not measures in the hemogram

Circulating pool (CP)- neutrophils flow freely in the center of (larger) vessels
CP is the only pool sampled by phlebotomy
CP = MP in dogs, horses, and ruminants
MP ~ 3X > CP in cats

Total blood = CP + MP
Average blood transit time is ~ 8-10 hours (all blood neutrophils replaced ~2.5x daily)
Tissue emigration random (NOT longest circulating leaves first), adhesion mediated, unidirectional, and “upregulatable” (inflammation)
Important- need to be in marginal pool to be in tissues (circulating → marginal → tissue)

98
Q

What is the difference between neutrophil kinetics in health versus in disease?

A

Health
Tissue survival ~1-4 days
Effete neutrophils (apoptotic) in tissues removed by macrophages

Disease

Tissue has purulent inflammation (pus) → most common cause of neutrophilia
Extravascular accumulation of neutrophils
Increased extravasation (endothelial activation and adhesion) and migration of neutrophils (chemotaxis) via inflammatory mediators towards a noxious stimulus

Inflammation → increased peripheral use, which results in
Decreased blood T1/2
Increased marrow release
Increased marrow production
Cytokine mediated

99
Q

Neutrophil kinetics regarding bone marrow (production and release)

A

Bone marrow release is first response
Increased marrow release mediated by G-CSF, IL-1, and TNF (inflammation) and corticosteroids
Manifests in blood rapidly, <1-2 days
Release is orderly - more mature released first
As mature neutrophils depleted (storage pool), younger cells are released → LEFT SHIFT
Indicative of depletion of mature storage pool
Typically in response to inflammation (increased demand) Therefore, left shift = inflammation but nor necessarily the converse (can get inflammation without left shift)
Neutrophilia +/- left shift is the usual response to inflammation
Neutropenia with left shift (degenerative) indicates “exhaustion” of marrow due (most often) to rapidly developing overwhelming infection (poor prognosis)

Increased effective granulopoiesis
Increased cell divisions at the myelocyte stage
Decreased attrition at myelocyte stage
Manifests in blood for 2-3 days (slower than increased release)

Stem cell input increase
IL-3 and GM-CSF increase the number of stem cells becoming myeloblasts
Total neutrophil production can be markedly increased, takes 3-5 days (longer first 2 mechanisms) to manifest in the blood - biggest effect

Rate of egress from blood
Inflammation results in decreased circulating T1/2 transit time
Corticosteroids increase circulating T1/2 (decreased adhesion)/transit time
Hypersegmented neutrophils (6 or more lobes) may be seen
Most common cause of hypersegmented neutrophils in blood, but there are other causes

Pool shifts
Shifts to circulating pool
Epinephrine
Corticosteroids
Shift to marginal pool
endotoxin

100
Q

Neutrophil kinetics regarding bone marrow (production and release)- species differences

A

Species with larger maturation/storage pools can mount larger responses

Dogs, cats, pigs = large storage pool species
Also rodents, birds, marine mammals

Horses, ruminants = small storage pool species
Neutropenia with left shift is common during early phase of (usually septic, purrelent) marked inflammation
Indicates serious disease but NOT marrow exhaustion
Hence, not as bad prognostically as same findings in large storage pool species (dog, cat, pig)
If an animal survives the first 2-3 days of disease, neutrophil count “recovers” to normal or neutrophilia

101
Q

With neutrophilia, what are events associated with physiologic leukocytosis?

A

Epinephrine mediated
Increased heart rate and blood pressure → displaces neutrophils → mild neutrophilia (2X) without left shift
Typically seen in excited, healthy young animals, especially cats and pigs

Excitement or exercise “fight or flight”

Marginal pool “shifts” to circulating pool
Total pool hasn’t changed but moved to marginal

Lymphocytosis may be present, especially in cats (may be up to 20,000/ microliter)

Neutrophilia most pronounced in cats because they have large marginal pools (3:1 ratio M:C)

Short term changes

102
Q

With neutrophilia, what are events associated with corticosteroid induced (stress reaction) leukocytosis?

A

Neutrophilia due to increased entry from storage pool, shift from marginal to circulating pool, and prolonged circulation time
Neutrophilia
Usually no left shift (unless depleted storage pool)
Lymphopenia (less marked in cats)
Difference in EPI induced → lymphophilia
Eosinopenia
Monocytosis (only in dogs)
Can be caused by exogenous or endogenous steroids
When sick, may induce this response
Can be seen in dogs with cushings disease → hyper adrenal corticosteroids
Occurs in hours-days while EPI takes minutes to seconds
May be seen concurrently with neutrophilia of other causes

103
Q

What is neutropenia and what are the causes?

A

Decreased blood neutrophil concentration
Patients with <500 neutrophils/microliter are considered at risk for sepsis

Causes
Decreased production of neutrophils
Could be due to viral infection or radiation and chemotherapy
No left shift
Increased demand
Increased demand in tissue than bone marrow can supply
Left shift
Peripheral destruction of neutrophils (immune mediated neutropenia)
Ineffective granulopoiesis
Common cause of myelodysplasia
Sequestration in the marginal pool → initial response to endotoxin

104
Q

Morphology in neutrophils- Toxic change causes

A

Leukergy- sticky leukocytes
Seen in horse with endotoxemia
Morphologic abnormalities of neutrophils and their precursors
Due to the presence of increased inflammatory mediators/cytokines +/- presence of bacterial toxins
Most often an indicator of “sepsis”/infection (esp. bacterial) when marked
EDTA can produce artifacts (time dependent) that complicate evaluation - “fresh” smear essential

105
Q

Morphology in neutrophils- Toxic change

A

Things seen
Döhle bodies (blue inclusions in the cytoplasm)
Diffuse cytoplasmic basophilia (blueness)
Cytoplasmic vacuolation (foaminess)
Abnormally large neutrophils (especially cats)
Persistent azurophilic or purple granules (“toxic granulation”) in the cytoplasm (rare finding most often seen in horses)

106
Q

Neutrophil Toxicity scale (based on morphology) and indication -

A

1+ - Döhle bodies only (spp. differences)
2+ - Döhle bodies + Diffuse cytoplasmic basophilia OR Cytoplasmic vacuolation
3+ - Döhle bodies + Diffuse cytoplasmic basophilia AND Cytoplasmic vacuolation +/- Toxic granulation
4+- Too toxic to differentiate from monocytes

Generally, 2+/3+ or 4+ toxic change indicates sepsis/endotoxemia
1+ to occasionally 2+ toxic change can occur secondary to ny strong marrow stimulation of granulopoiesis (immune mediated hemolysis, necrosis, etc)

107
Q

What does neutrophil toxic change indicate?

A

Morphological changes indicate increased cytoplasmic contents (ribosomes, endoplasmic reticulum) and lysosomal enzymes and/or bacterial substances denaturing/damaging cytoplasmic proteins and organelles
Compromised function?

108
Q

Morphologic changes- intracytoplasmic organisms causes

A

Bacteria in circulating neutrophils of septicemic animals
Uncommon
Poor prognosis: overwhelming infection

Intracytoplasmic organisms- specific infections, specific diseases
Can recognize on routine hematology

109
Q

What is eosinophil morphology?

A

Bright orange granules in cytoplasm
Staining is due to their highly basic (cationic) proteins, predominantly MBP and ECP
Not recognizable before the promyelocyte/myelocyte stage

110
Q

What are eosinophil species differences?

A

Eosinophil granules vary in size and shape between species
Horse- most distinctive with giant, bright granules

Feline- rice shaped and rod shaped granules → looks like a heterophil

111
Q

What is involved with eosinophil production and kinetics?

A

Develop from distinct progenitor in marrow, CFU-Eo (colony forming unit- eosinophil)

Variable time for development (2-6 days)

IL-3, GM-CSF, and IL-5 are the major players
IL-3 and GM-CSF are for all leukocytes
IL-5- specifically stimulates eosinophils

A maturation/storage pool exists
Marrow reserve variable (spp. Specific - eg. guinea pig has marrow:blood ratio of 300:1 versus 3.4:1 humans)

Blood T1/2 very short (species variable)

Marginal pool exists

Tissue life span longer than neutrophils (6 days +) and can be increased under the influence of cytokines
Don’t recirculate (like neutrophils)

Eosinophils preferentially reside in skin, GI tract, and lung (blood:tissue ~1:200-300)

Adherence and emigration through endothelium occurs via similar mechanisms to neutrophils, but different set of chemotactins
Chemokines, esp eotaxin
antigen/antibody complexes- chemoattraction
ECF-A (from mast cells)- downregulating mast effect
Histamine- mast cells are full of it
LTB4- arachidonic acid metabolites
C5a, C567- complement proteins
PAF- platelet attractant factor
Parasite and damaged tissue products

112
Q

What are eosinophil functions?

A

Helminth killing (+Ab + complement = opsonization)
Type I hypersensitivity / allergic reactions (regulatory role)- phagocytosis immune complexes and “neutralize” mast cell products (histamines)
Phagocytosis and killing of bacteria (Very minor)

113
Q

What are some mechanisms that cause eosinopenia?

A

Corticosteroids (main cause)
Part of the “stress” leukogram
Corticosteroids may attenuate or dampen eosinophilia of other causes
Ex. there are always low concentrations of histamine in blood all the time and steroids stop Eo from coming out

114
Q

What are some mechanisms that cause eosinophilia?

A

Parasitism (helminth parasites)
Allergies or hypersensitivities- esp. Skin, gut, lung locations (IgE, IgG leads to mast cell granule release: chemotactic to Eos)
+/- fungal disease (esp. horses)
“Sensitized” T cells produce a more marked eosinophilia upon second exposure

Paraneoplastic
Mast cell tumors
Spontaneously degranulate → eosinophilia attracted to it
Lymphoma (especially T cell)
Many other
Eosinophilic leukemia is rare

115
Q

What is basophil morphology?

A

Easily recognizable with blue purple granules
Staining due to highly acidic (negatively charged) proteins in secondary granules
High affinity for basic dyes (basophilic color)
Histamine, heparin, sulfated mucopolysaccharides
Granules typically fill cytoplasm but less numerous in the dog, “paler” in the cat
Maturation parallels that of neutrophils and they have a similar nuclear morphology
Not recognizable before the promyelocyte/myelocyte stage

116
Q

What are species differences in basophil morphology?

A

Dog- not many granules

Horse- very granulated (dark purple)

Cat- pale, lavender colored granules

117
Q

What is associated with basophil production and kinetics?

A

Distinct marrow progenitor- CFU-Baso
Similar cytokines as involved with eosinophil production (IL-3, GM-CSF, IL-5). Marrow storage minimal
Kinetics poorly understood - blood half life ~ 6 hours, tissue lifespan 2 weeks?

Inverse relationship between blood basophil counts and tissue mast cell numbers (ie. cats versus rabbits)
Cats- lots of blood basophils and not many mast cells
Rabbits are opposite

118
Q

What is basophil function?

A

Similar contents to mast cells and likely similar functions
Degranulation secondary to antigen binding and crosslinking surface Ig (release of inflammatory mediators)
Only known source of heparin and activators of lipoprotein lipase
Basophils go up when there are changes to lipid metabolism

119
Q

What are the causes of basopenia?

A

Not clinically recognized as so few present normally

120
Q

What are the causes of basophilia?

A

Uncommon, usually mild
Often accompanies eosinophilia
Lipid disorders, feline heartworm disease, myeloid neoplasms (mechanism unknown)

121
Q

What is monocyte morphology?

A

Largest cells in the blood (on a blood smear)
Artifact of smearing → likes to stick to glass
Variably shaped, “ameboid nuclei” with abundant, blue-gray cytoplasm, often vacuolated
Precursors difficult to ID- monoblasts present in very low frequency in marrow and difficult to distinguish from myeloblasts with routine stains
Increased vacuolation may indicate activation (ddx artifact due to excessively long EDTA storage)
Becomes activated → macrophage in tissue

122
Q

What is associated with monocyte production and kinetics?

A

Monocytes are immature macrophages, capable of mitosis, undergo further differentiation
Develop from bi-potenital progenitor cell (CFU-GM)
Major factors are IL-3, GM-CSF, and M-CSF (specific)
Marrow production is rapid (2-3 days)
No marrow storage pool (blood is the storage pool)
Blood circulation time longer than neutrophils ~24 hours
Marginal pool exists in some spp. (human, mouse, dog, rabbit)
Leave blood randomly, don’t recirculate and become macrophages in the tissues- “fixed” and “wandering” (mesothelial, joints, lung)
Long tissue T1/2 and can divide in the tissues

123
Q

What are the functions of monocytes?

A

Immature macrophages

Macrophage functions:
Phagocytosis
Antigen processing and presentation
Cytokine production (inflammation, hematopoiesis)
Destruction of effete cells and debris
Pinocytosis and catabolism of plasma proteins
Specialized “fixed” function ie. Kupffer cells

124
Q

What are the causes associated with monocytosis?

A

Present with inflammation
Monocytosis has correlation, albeit relatively poor, to tissue macrophage accumulations (chronic inflammation)
Monocytosis may occur during acute purulent inflammation (esp. If there is associated tissue destruction) as well as chronic inflammation
Corticosteroids cause monocytosis, esp. Dogs, less so other spp.

Response to disease, secondary to
Persistent inflammatory stimuli- viral, fungal, atypical bacterial infections
Component of steroid-induced leukogram (primarily dogs)
Diseases with increased tissue demand for macrophages
Immune mediated diseases, necrosis, malignancy, hemolysis, pyogranulomatous disease

125
Q

What are the causes of monocytopenia?

A

No clinical significance

126
Q

What is lymphocyte morphology?

A

Usually the smallest and most featureless of the blood leukocytes
Typically have a high N/C ratio with sparse cytoplasm that is variably blue
Lymphocytes are often the predominant leukocyte in the peripheral blood of ruminants (in health)
Activated lymphocytes are larger
Can be granular - seen in dogs and cats

Differentiation of lymphocytes is manifest by acquisition and loss of lineage specific cell surface antigens (CD antigens)
T cells
B cells
Morphologically, it is usually not possible to determine to what subpopulation and stage of differentiation a lymphocyte belongs

Plasma cells are recognizable as terminally differentiated, Ig secreting B cells

127
Q

How does one differentiate between the different types of lymphocytes? How do you differentiate between the different maturation stages?

A

Using CD markers and testing

128
Q

What are the kinetics of lymphocytes?

A

Blood lymphocytes originate and recirculate from numerous sites (distribution and relocation)
Thymus
Marrow
Lymph nodes
Spleen
MALT

Recirculation mechanisms complex and involve molecules we’ve already discussed
Selectins, integrins, ICAMS, chemokines, lymphatics, and HEVs

Recirculation is crucial and facilitates
Generalized distribution
Re-location (immune surveillance)

Many lymphocytes are long lived cells

Recirculation is common via lymphatics and specialized vasculature (HEVs)

Appearance may change under influence of antigenic or cytokine stimulation (become “reactive” when stimulated)

The majority of circulaitng lymphocytes in most species are T cells
~70% T, 20% B, 10% NK

Blood lymphocyte numbers have relatively poor correlation to tissue lymphocyte numbers
Most lymphocytes are in tissue

Blood lymphocyte counts are higher in young, growing animals and decrease with age
Younger animals encounter many novel stimuli

129
Q

What is lymphocyte function?

A

Ig production- B cells
Cytokine production (predominantly CD4+)- T cells
Cytotoxicity (CD8+, NK cells)

130
Q

What are the causes of lymphopenia?

A

More common finding on CBCs of sick animals than lymphocytosis

Corticosteroid mediated (usually)
Lymphocytes apoptosis in response to corticosteroids
Get hung up in tissue and not as much recirculation

Depletion
Lymph loss (chylothorax, lymphangiectasia, inflammatory or neoplastic intestinal disease resulting in lymphatic obstruction)

Lymphoid hypoplasia/aplasia
Hereditary immunodeficiency (CID or arabian foals), acquired immunodeficiency, thymic aplasia/atrophy

Immunosuppressive therapy, radiation

Acute systemic infections- esp. Viral infections
K9 distemper, K9 parvovirus, FIP

Disruption of normal lymph node architecture
Prevents recirculation eg. lymphoma, granulomatous disease (lymphadenitis)

131
Q

What are the causes of lymphocytosis?

A

Young animals have higher counts - may be out of adult reference interval

Epinephrine - mediated (physiologic), esp. Cats
Due to increased BP → increased lymphatic pressure
Thoracic duct empties → lymphocytosis

Immune stimulation (fairly rare situation - typically increases numbers, but stays within reference interval)

Persistent inflammation
Increased lymphocytes as a result of increased lymphopoiesis, lymph node hyperplasia can be noted
Also called “reactive” lymphocytosis (antigenic stimulation, chronic infection)
Considered viral, atypical bacterial, fungal, rickettsial, protozoal infections among others (“persistent” infections)
Concurrent hematologic abnormalities may include mature neutrophilia and monocytosis

Neoplasia
Neoplastic proliferation of lymphocytes in marrow, lymph nodes or other lymphoid tissue
Not all lymphomas are associated with lymphocytosis
In fact, most are not!!
Persistent lymphocytosis in cattle
subclinical , non neoplastic manifestation of BLV infection in cattle

132
Q

What are morphologic abnormalities in lymphocytes?

A

Reactive lymphocytes
Immune stimulation, including infections

Atypical lymphocytes (neoplasia)

Plasma cells

133
Q

What are the principles of antigen recognition by B cells and T cells (immune protection)?

A

Recognizes specific epitopes on antigens that they did not encounter during development (“non-self”)
B cell- BCR can bind to an epitope on many different types of antigens: proteins, lipids, carbohydrates, nucleic acid (DNA), and it can be either
It may bind to a stretch of amino acids (continuous or LINEAR epitope)
OR it may bind to a particular tertiary or quaternary structure (conformation) on an antigen = CONFORMATIONAL epitope

T cell- the majority of T cells recognize peptide epitopes
Small subsets of T cells
Lipids (NKT)
Metabolites (MAIT)
T cells bind a particular amino acid sequence. These are linear epitopes in the context of an antigen-presenting cell
Activating a naive T cell- DC
Activated T cells- lots of cells

134
Q

What is the basic structure of immunoglobulins, the B cell receptor (BCR)

A

B cell- 2 light chains and 2 heavy chains and associated with signaling chains
Heterodimer associated with two signaling chains
Variable region- light and heavy chains
Constant region- heavy chains

135
Q

What is the basic structure of immunoglobulins, the T cell receptor (TCR)?

A

T cell- TCR resembles a membrane bound Fab-fragment
2 variable
2 constant regions
Encoded by T-cell alpha and beta or gamma and delta chains
Heterodimeric protein linked by a disulfide bridge
Each chains has a variable (V) and constant (C) region
Highly homologous to immunoglobulin
TCR has one antigen binding site
TCR is never secreted

136
Q

What is the relationship between B cell receptors and antibodies (diagnosis, immune protection)?

A

The BCR defines specificity of a B cell for its “cognate antigen”
Every B cell develops one distinct BCR that when secreted by the cell is called an antibody

137
Q

What is the difference between CD4+ and CD8+ T cells?

A

CD4- MHC II (class II restricted)
CD8- MHC I (class I restricted)

138
Q

What is the function of MHC I and MHC II (“transplantation antigen”)?

A

MHC are highly variable and present varying peptides to T cells
I- can fit 8-10 amino acids long
II- can fit 13-25 amino acids long

139
Q

What are the principles of B cell lymphocyte development? Failure results in immunodeficiencies

A

B cells- Class switch recombination
Process of generating the Ig is recombination

Requires recombination activating gene (RAG1 and RAG2), and DNA repair enzymes
Defects in these enzymes → severe combined immunodeficiency (SCID) as B- and T-cells develop

A diverse repertoire of due to multiple genes encoding the variable region
Complementarity determining regions (CDR)
CDR3: region that is hypervariable
Recombination + Non-genome encoded nucleotides

140
Q

What are the principles of T cell lymphocyte development? Failure results in immunodeficiencies

A

Selection of T cells

T cell progenitors from the bone marrow home to thymus → enters the corticomedullary junction → cortex → medulla → exit to periphery (as a single positive- only CD4+ or CD8+)

In the cortex, Cell is DP (double positive) and through initial TCR:MCH interaction (in thymus), it can be either CD4+ or CD8+

Can also be double negative → apoptosis

In the medulla, antigen presented by epithelial cells (medullary), or dendritic cells (DC)
Interaction leads to removal from body

Aire is a transcription factor that allows the transcription of tissue antigens by medullary thymic epithelial cells (mTEC)
If no interaction, mature cells are released from the thymus into blood

141
Q

How does the selection of B cells work

A

Selection of B cells- three checkpoints for rearranged Ig chains for functionality and self-reactivity

Pre-B cell: Is heavy chain functional? Expression a “pre-BCR” on the cell surface consists of VDJH-IgM constant region, surrogate light chains (VpreB, Lamda5), CD79a/b BCR signaling chains
Outcomes
Weak signal- cell proliferate and undergo light chain rearrangement
Signaling too strong- cell is eliminated (reacts with antigens in bone marrow)
No signaling- cell undergoes apoptosis (cannot signal)

Immature B cells: is the BCR functional? Surface expression of the BCR
VDJh-IgM constant region and VJL kappa light chain fully rearranged
CD79a/b BCR signaling chains
Outcomes
Weak signal- cells proliferate and leave bone marrow
Signaling strong- cell undergo rearrangement on lamda light chain locus
Signaling too strong- cell undergoes apoptosis (reacts too strongly with antigens in bone marrow)

Spleen- transitional B-cell: is the BCR reactive to antigens in the spleen?
VDJh-IgM constant region and VJL kappa light chain fully rearranged
Start to have IgD constant region BCR replace the IgM
CD79a/b BCR signaling chains
Outcomes
Weak signal- cells fully mature resting B cells
Signaling too strong- cell undergoes apoptosis (reacts too strongly with antigens in the spleen)

142
Q

What are the principles of central and peripheral tolerance? Failure results in antibody-mediated autoimmune diseases

A

Selection process eliminates B cells that recognize self-antigens “self-reactive”
Central tolerance- in the bone marrow
Peripheral tolerance- in the spleen

143
Q

Why are blood groups clinically important?

A

Allows for safe administration of compatible transfusions
Also allows for avoidance of neonatal isoerythrolysis

144
Q

What are blood groups? What is important in understanding when it comes to transfusion testing?

A

Blood groups defined by genetically determined markers (known as antigens) on the surface of erythrocytes
Varies from species to species
If it has the marker, it is positive
If it doesn’t have the marker, it is negative

The markers are considered “antigenic” (recognized as non self) in individuals lacking the same marker
Antigenicity- likelihood that the immune system will react and make antibodies against the foreign substance
If we give wrong donor blood to a transfusion recipient → can result in hemolytic transfusion reactions
Surface antigens + opposing antibodies ⇒ agglutination (clumping) and hemolysis
Important to know which blood types the donor and recipient are and which blood types and circumstances are likely to cause immune reaction

Pre-transfusion testing minimizes the risk of incompatible transfusions and minimizes exposure of transfusion recipients to foreign red cell antigens they lack to prevent new antibody formation
Blood typing- identifies the primary rec cell surface antigens of the donor and recipient
Crossmatching- identifies any blood groups incompatibilities
Major- tests for alloantibodies in recipient’s plasma against donor cells
Minor- tests for alloantibodies in donor plasma against recipient’s RBCs

145
Q

Dog blood groups and implications

A

estimated 13 blood types

DEA (dog erythrocyte antigen)
DEA 1 most important → strong alloantibody response after sensitization → most clinically significant to test for
DEA 3, 4, 5, 6, 7, 8

Others
Dal
Kai-1, Kai-2

Dogs lack clinically important natural alloantibodies pre-sensitization
DEA-1 has no naturally occurring alloantibodies
Variable naturally occurring antibodies in other DEA negative dogs (3, 5, 7)
No known natural antibodies to Dal or Kai-1/Kai-2
First transfusion → cross matching is not required but recommended, especially if IMHA
If repeat transfusion (or unknown history), required cross matching for transfusion

DEA 1- donor → recipient can be DEA 1- or DEA 1+
DEA 1+ donor → only to DEA 1+ dogs

146
Q

Cat blood types and implications

A

AB blood group system
Type A (majority of DSH/DLH and non pedigree cats)
Type B (uncommon but varies with breed/location)
Type AB (rare)

MiK antigen
Patients are positive or negative
Most DSH have MiK

Pre-existing alloantibodies
Type A: weak anti-B antibodies
Type B: strong anti-A antibodies
Type AB: no A or B antibodies
Mik negative cats: anti-Mik antibodies

Cats should be typed and cross matched before transfusion- confirm type B or AB due to rarity
Potential for hemolytic transfusion reactions on first transfusion

Transfuse
Type A → type A cats
Type B → type B cats
Type A → type AB cats because there are not many AB donors

Neonatal isoerythrolysis
Litters from type B queens (+type A toms)
Type A or AB kittens are at risk
Kittens receive anti-A alloantibodies through colostrum → hemolysis
Contributes to “fading kitten syndrome”
Prevention
Blood type prior to mating
Remove kittens from type B queens for first ~24 hours

147
Q

Horse blood types and implications

A

8 major blood group systems (7 internationally recognized)
A, C, D, K, P, Q, U, (T)

> 30 different RBC antigens (factors) recognized within these groups
Type-specific blood transfusions nearly impossible
Aa, Ca, and Qa most important clinically for their role in hemolytic transfusion reactions and neonatal isoerythrolysis

naturally occurring alloantibodies → ~90% do not have them, but ~10% do to Aa and Ca
Antibodies can develop after transfusion or pregnancy (most common for Aa and Qa)

There is no universal donor so attempt to match donor and recipient types as closely as possible
Especially A, C, Q
Other testing options
Donors should have negative antibody screen
Crossmatch

Testing limitations
Access to donors and rapid typing?
Mostly Aa and Ca positive donors (majority of horses have this blood type)

Emergencies
First transfusion might be performed without compatibility testing
If typed donors not available → use healthy gelding of same breed
Usually well tolerated, unknown RBC lifespan

Neonatal isoerythrolysis
Higher incidence in
Foals born to Aa or Qa negative mares
Some breeds (eg. Friesans)
Mule foals due to “donkey factor”
Prevention
typing/antibody screening
Muzzle foal for 24-48 hours + alternative colostrum

148
Q

Other animals’ blood types and implications

A

Matching of blood types is not practical
First transfusion risk- typically minimal
Choose blood donor of same species transfusion
Crossmatch if high risk

149
Q

What is the function of dendritic cells in T cell activation? What is its bridge function?

A

Dc are professional antigen presenting cells

Uptake antigen from peripheral sites and migrate to the afferent lymphatics
Guided by chemokines
DC can be activated by many types of PAMPs/DAMP
Presentation in the lymph node activates adaptive immune responses
Therefore, DC are a bridge between adaptive and innate immunity

150
Q

What are the steps required that lead to the activation of CD4 and CD8 cells? For DC

A

If activated by PAMPs/DAMPs or pro-inflammatory cytokines,
endocytosis /environmental sampling is reduced
Increases expression of chemokine receptors
Migrate towards afferent lymphatics
Increase expression of cell surface proteins
Migrate to T cell zones
Present antigens by MHC I and MHC II (MHC I: antigen is turned into smaller fragments → kept in ER; MHC II: fuse of phagolysosome and goes out into MHC II)
Activate antigen-specific CD8 and CD4 T cells

151
Q

What are the differential requirements for CD4 and CD8 cells in antigen presentation via MHC II (exogenous antigens) and MHC I (mostly endogenous) peptide complexes, in response to vaccination?

A

Signal 1- antigen specific stimulation
Ag peptides/MHC complex
TCR interacting with MHC
In naive T cell, this isn’t enough to activate

Signal 2- Co-stimulation
Membrane bound proteins
CD28 on T cells
CD80/CD86 (aka B7.1/B7.2) on DC

Signal 3- polarization
Determine the “character” of effector mechanism
Induce correct gene transcription to deal with problem at hand
Membrane bound molecules
Cytokines - “instructions” with how to deal with this antigen and tells it where to go

152
Q

What are the major functions of CD4 and CD8 T cells?

A

CD4+- T helper cell
Makes cytokines
Can be either T follicular helper cells or T regulatory (induced)
Different signal 3’s can give rise to different effects and cytokines produced

CD8+- cytotoxic
Punch holes (using perforin) in target cells and enzyme goes in there and kill the target cell

153
Q

What are the different signals that lead to the activation of CD8 T cells and NK T cells causing cell lysis?

A

NK cells- kill host cells that lack MHC I
Acts as an inhibitory signal in the NK cell
Not antigen specific
Often consequence of viral infection
Some viruses decreases MHC I presentation → no more stop signal → cell death

CD8 cells- recognition of antigen:MHC I complex
Antigen specific killing → stimulation to kill

154
Q

What are the activation requirements and the functions of naive and memory T cells?

A

Naive cells- function as pool for activation
Activation requires all three signals

Memory T cells- after initial response, waiting to see antigen again to mount response
Effector T-cells die off after insult is cleared
Three main types of memory
Tcm- central memory (reside in LN, spleen, circulate)
Tem- effector memory (circulate in/out of tissues)
Trm- Tissue resident memory (reside in tissue, site of insult)
Antigen-experienced CD4+ or CD8+ T cells that can be reactivated by “signal 1” alone
Increase the number of antigen-specific T cells for faster and stronger recall responses

155
Q

Describe the life cycle of platelets

A

Following release of pre-platelets → mature platelets
30% of platelets storage in spleen
Life span: ~4-6 days

Sensensence leads to desialylation
Loss of sialic acid exposes galactose which triggers
Phagocytosis by Kuffer cells
TPO production

156
Q

Describe the regulation of platelets

A

Regulation → mediated by hormone, thrombopoietin (TPO), which is made in the liver primarily (kidneys, smooth muscle)
Levels of TPO are regulated by platelet numbers (“sponge theory”)
Platelets express receptors that bind to TPO and internalize TPO
TPO does not cause fragmentation of proplatelets from MK

Other stimulants of TPO
Senescent “aged” platelets
Inflammatory cytokines
IL-6
IL-11
GM-CSF
CLL5

157
Q

Describe the process of thrombopoiesis

A

Sites of platelet formation
Bone marrow
Bloodstream
Lung

Megakaryoblasts undergo endometriosis (endoreplication) to become polypoid
Endomitosis- all the steps of mitosis except they don’t split

Each MK releases ~10^4 platelets

Production of all necessary membrane proteins, granules, organelles, and genetic material (RNA) for a mature platelet to survive circulation

MK extends long cytoplasmic processes with platelet-sized beads, known as proplatelets
Proplatelets further undergo fragmentation to form mature platelets in circulation

158
Q

Identify structures and pathways in platelets that are crucial to clot formation.

A

Platelet membrane- fluidity of phospholipid bilayer maintained by cholesterol, phospholipids, and influenced by temperature

Receptors “float on” the bilayer and act in concert when given the signal
When temperature <15 to 18 degrees celsius, fluidity of membrane declines (phase transition)
Gel consistency → R can’t move on cell membrane → causes isolation or crash together (too little or too much signaling)
This shift activates platelets inappropriately or decreases their ability to respond

Phospholipids on platelet membrane are heterogenous
Inactive membrane- neutral phospholipids are located in the exterior while electronegative phospholipids face the cytoplasm
Electronegative phospholipids are “flipped” when platelets are activated

Integrins are heterodimeric (alpha and beta subunits) transmembrane receptors linking to the cytoskeleton
Crucial role in platelet plug formation and clot retraction
Integrin alpha IIb beta 3 (pka GPIIb/IIIa)
In unstimulated platelets, they remain in “low affinity” state
Platelet agonist activated the integrin in the cytoplasmic tail (beta3) → opening of ectodomain → “high affinity” state

159
Q

What is the 3-stage model of platelet activation during hemostasis.

A

initiation
extension
stabilization

160
Q

Describe platelet activation initiation

A

platelet adhesion to damaged endothelium
Exposure of collagen and release of vWF (von Willebrand factor) from endothelium
vWF cleaved and immobilized on collagen fibers
Platelets bind to vWFs (tethering and rolling)
Facilitates formation of initial platelet monolayer

161
Q

Describe platelet activation extension

A

activation of platelets due to adhesion to the monolayer. Release of agonist for self-activation and recruitment of additional circulating platelets to the initial monolayer

Secretion of…
ADP
Binds to P2Y12 and P2Y1 → causes Ca mobilization, TxA2 generation, and platelet aggregation
Inhibits Prostaglandins
Sources- denes granules, damaged cells at vascular site of injury, breakdown of ATP, works synergistically with other platelet agonists
P2Y12 can be inhibited by a class of drug called thienopyridines (clopidogrel, prasugrel)

Thromboxane A2
Arachidonic acid is cleaved by the enzyme, phospholipase A2 (PLA2), activated by many other agonists
TxA2 is generated from the fatty acid, arachidonic acid (AA), in the cell membrane via COX-1
TxA2 generated can self-activate platelets or activate nearby platelets via TR
Aspirin inhibits platelets by inhibiting COX-1 decreasing generation of TxA2
Non-steroidal anti-inflammatory drugs (NSAIDs)

Thrombin
Thrombin is generated via secondary hemostasis
Most potent activator of platelets
Thrombin activates protease activated receptors (PAR) on platelets
Strong couple of phospholipase C resulting in profound elevation of intracellular Ca++
Granules are also secreted during activation

Platelet shape change
Occurs secondary to activation or changes in blood flow (shear stress)
Transformation from biconcave disk to fully spread cells
Increases surface area to facilitate further adhesion to site of vascular injury and other platelets
Must occur prior to platelet aggregation

Formation of procoagulant membrane
Membrane is heterogeneous
Platelet activation externalizes electronegative phospholipids
They provide “docking” surface for coagulation factors to form complexes for thrombin generation (Factor II)

Integrin activation

162
Q

Describe platelet activation stabilization

A

Binds to extracellular matrix proteins
Activation of alpha IIb beta3 → fibrinogen, fibronectin, and vWF
Platelet aggregation- platelet to platelet interaction through fibrin mesh
Once alpha IIb beta3 binds to its ligand, platelets undergo “final activation process”
Clot retraction and stabilization of the platelet plug

163
Q

What is the role of platelets in secondary hemostasis?

A

Thrombin is generated by secondary hemostasis
Alpha granules released from platelets are important for secondary hemostasis
Facilitates fibrin formation

164
Q

What are the differences between primary versus secondary hemostatic defects?

A

Primary- like letting the tap drip slowly, so symptoms are
Petechiae
Ecchymosis
Epistaxis
melena/GI
Hemorrhage (dog)
Scleral hemorrhage

Secondary- like not turning off the tap at all
Hematomas
Hemoabdomen
Hemothorax
Hemarthrosis

Both could be due to trauma or surgically induced
Primary- platelets, endothelium, vWF
Secondary- coagulation factors, fibrinolysis (removal of clots after formation)

165
Q

How can platelet count be assessed on peripheral blood smears?

A

Mean platelet volume (MPV)- the mean volume of all platelets
Large platelets in circulation due to increased fragmentation and increased demand (higher TPO)
Indication of active bone marrow response

Manual count- count # of platelets in 10 100x oil immersion field monolayer
Estimated platelet count X 10^3/ul = average # of platelets in 10 fields x 15
If there are clumps, estimated count should be considered a minimum value

166
Q

What are the major causes of thrombocytopenia? What are some differential diagnoses for each cause?

A

Low platelet count- can be either artifact/pseudothrombocytopenia, macrothrombocytopenia, or thrombocytopenia

Artifact
Dilution
Clumping due to in vitro platelet activation and aggregation
Increased duration from sampling to anticoagulant → clot formation
Inadequate mixing of blood and anticoagulant
Poor venipuncture

Pseudothrombocytopenia
Anticoagulant (EDTA)-mediated alteration in integrin causing agglutination by antibodies, IgG or IgM
Use other anticoagulants like heparin or citrate (account for ~10% drop in platelet count for citrated samples)

Macrothrombocytopenia - breed dependent mutations in beta 1 tubulin leading to “Giant Platelet Disorder”
Defective fragmentation of proplatelets from megakaryocytes
Characterized by
Low platelet count (as low as 20,000/ul)
High MPV
No bleeding diathesis
Normal function, no clinical signs in dogs

Thrombocytopenia

Increased consumption
Acute onset of hemorrhage → loss +/- consumption of platelets from trauma or surgery
Disseminated intravascular coagulation (DIC)- ongoing consumption of coagulation and platelets due to underlying diseases
Snake envenomation
Moderate to marked thrombocytopenia

Increased destruction- immune mediated thrombocytopenia (ITP/IMTP)
Platelets are phagocytized by splenic/hepatic macrophages mediated by anti-platelets antibodies and complement
Primary is due to spontaneous autoimmune disease, while secondary is due to underlying disease
Marked thrombocytopenia → spontaneous hemorrhage

Decreased production
Thrombopoiesis affected at the level of the bone marrow
Myelophthisis caused by fibrosis, neoplasia, or granulomas
chemotherapy, radiation, viral infection (canine parvovirus, feline panleukopenia virus)
Other types of blood cells are likely affected
Moderate to marked thrombocytopenia

Increased sequestration
Spleen harbors up to 30% of circulating platelets (seen in splenomegaly)
endotoxemia/sepsis → platelet activation → sequestration in pulmonary vasculature or other microvasculature
Thrombocytopenia is mild

167
Q

What are the major causes of thrombocytosis? What are some differential diagnoses for each cause?

A

increase in number of platelets in circulation (>500 x 10^3 /L)

Primary- when idiopathic, while secondary- from other diseases

Inflammation, neoplasia, endocrine (hyperadrenocorticism)

168
Q

What molecular structures in platelets are crucial to thrombus formation? How can platelet disorders lead to bleeding diathesis?

A

Initiation- platelet adhesion to damaged vessels
vWF and exposure of collagen is important
Platelets express receptors that bind to collagen and vWFs

169
Q

What are some commonly available platelet function assays?

A

Tier 1 Tests

Template bleeding time (TBT)- making cuts in the animal (standardized) and recording the time it takes to stop bleeding

Buccal mucosal bleeding time (BMBT)- standardized cut into lip and recording time it takes for bleeding to stop
Crude assessment of initiation phase of coagulation

Platelet function assay- 100/200
Cartridge based assay that mimics in vivo high-shear environment. Whole blood flown through an aperture and membrane coated with collagen +/- ADP or epinephrine
Time takes for aperture to close measured as “platelet closure time”
Assess vWFs and platelet function
Highly automated with high precision and repeatability
Not widely available and non-specific (screening test)

Quantity- immunodetection using anti-vWF antibodies (vWF:Ag)
Patient plasma compared to species-specific pooled plasma from healthy animals
Diagnostic = vWF:Ag < 50%
Equivocal or borderline = vWf:Ag 50 to 70%
Normal = vWF:Ag > 70%

Multimeric assay
Analysis of multimers by separation of vWFs based on molecular weight by electrophoresis and immunoblot
Rarely performed in clinical patients

Functional assay
Rarely performed in clinical patients
vWF to collagen binding (vWF:col)

For known mutations
Genetic testing- test for potential carriers, patients with equivocal vWF:Ag

Tier 2 Tests
Aggregometry
Collects platelet rich plasma and measures light that can pass through

Flow cytometry
Characteristics: cell size/granularity
Quantity specific population of cells
Expression of cell membrane receptors
Intracellular protein expression
Intracellular protein phosphorylation

170
Q

Explain B activation regarding T dependent/T independent

A

T independent- non protein antigens can activate B cells
Typically structures with repeating epitopes
Polymers, carbohydrates, lipids

T dependent- most effective B cell responses, supported by CD4+ T cells
After activation and clonal expansion,
Migration to B-T cell border
Receive signals (CD40 ligand on T cell)
Differentiate into long lived plasma cells or memory B cells
Step 1- antigen binds to BCR
Step 2- antigen is internalized and processed for presentation on MHC II
Step 3- B cell with epitope on MHC II interacts with T cell that recognizes that specific antigen
B cell co-stimulation signaling: interaction of T cell CD40L and CD40 on B cell
Cytokines from T cell skew or induce isotype in the B cell

171
Q

Explain B activation steps and procession

A

Antigen goes into afferent lymphatics → in the subcapsular sinus, complement receptor binds to it from subcapsular macrophage → B cells pass it along to each other until it gets to the B cell follicle → passes antigen to follicular dc → mature B cells see if it can bind to antigen epitope → proliferates if it does → goes out efferent lymphatics

Similar process in the spleen → passing of antigen in marginal zone until it gets to follicular dc in the primary follicle

Naive B cell → activation (binding to antigen) → clonal expansion → activated B cells migrate to T-B border zone

Development of B cell (VDJ) → somatic hypermutation (random point mutations in V region) → class switch (constant regions)

172
Q

Explain B activation regarding Consequences of activation

A

Prior immunization increases the number of specific B and T cells

Some of these will become memory B and T cells
Respond faster to BCR and TCR activation
No need for costimulation to activate memory T cells
Memory B cells rapidly differentiate in an extrafollicular response = fast production of antigen specific class-switched antibodies

Challenge (re infection or booster immunization) gives faster and stronger responses
This is the basis for vaccine induced immune protection

173
Q

Explain B activation regarding Basics of class switch recombination

A

Activation induces “Activation-induced deaminase” (AID) expression

Switch the Ig heavy chain constant region → produce different Ig isotypes
Loops different parts → parts that aren’t in loop → constant that influences Ig type

This is based on cytokine signaling

Constant region dictates structure → structure lends to function (# of binding sites)
Receptors for these isotypes are differentially expressed and may have unique affinities for each isotype
Gives rise to what it can bind

In early response, Ig is mostly IGM, but as time as goes on, more class switching → more IgG response

174
Q

Explain B activation regarding T cell dependent activation producing Short lived extrafollicular plasmablasts

A

T independent- after activation, B cells differentiate into antibody-producing plasma cells → short lived cells
At border of B-T cell zone
Cells reside in medullary cords of LN or red pulp of spleen
Rapid generation (days-weeks)
Generation of short-lived plasma cells
No/little memory

175
Q

Explain B activation regarding T cell dependent activation producing Germinal center formation (inside B cell follicle)

A

T dependent
Lymph tissue follicles
Develop slowly (1-2 weeks)

Introduction of point mutations in the variable region that encodes BCR “somatic hypermutation”
Changes BCR - random point mutations that are most frequent in the VDJ region
Since random, some BCR will have low affinity to helper T → apoptosis
Some will have high affinity → T cell helps B cell mature and proliferate → memory B cell and plasma cell

Generates long-lived high-affinity plasma cells that migrate to bone marrow

Leads to memory B cells

176
Q

What are the functional differences between memory B cells and plasma cells?

A

Memory
Do not secrete antibodies
Circulate between blood and secondary lymphoid tissues
Can reside in other tissues
Often Ig class switched (not IgM+)

Plasma cells
Migrate to bone marrow
Secrete class switched and high affinity antibodies into the blood
Antibodies persist for months to years in serum and plasma

177
Q

What are the principles of ELISPOT

A

clinical test for exposure to antigen by determining frequency of antigen specific IFNg secreting T cells
Using the whole blood from patient, which contains APC and CD4+ T cells

Add antigen to the cells
Antigen phagocytosed by APC
Presented to T cells by MHC II
On plates that are coated capture with antibodies that bind IFNg
After detection, antibody and substrate addition, count stops

178
Q

What are the principles of ELISA?

A

Antibody capture (indirect ELISA)
Antigen coated well → addition of patient serum → specific antibody binds to antigen → wash and enzyme linked antibody binds to specific antibody → wash and substrate is added and converted into colored product and measured with absorption

Antigen capture (sandwich ELISA)
Monoclonal antibody coated well → wash and addition of patient serum → antigen binds to antibody → wash and second monoclonal antibody, linked to enzyme, binds to immobilized antigen → wash and substrate is added and converted by enzyme into colored product and measured as absorption