• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/429

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

429 Cards in this Set

  • Front
  • Back
  • 3rd side (hint)
T/F: Lymphocytes, Phagocytes and Mediator cells are all derived from hematopoietic stem cells
TRUE
Whats the difference between primary lymphoid organs and secondary lymphoid organs?
Primary = where immune cells ARISE (bone marrow, thymus)

Secondary = where immune cells come together to initiate adaptive immunity (lymph node, spleen)
What are the three major functions of the immune system?
Defense, Homeostasis, Surveillance
Define Defense, a function of the immune system
prevent disease caused by microbes, viruses, parasites, fungi, bacteria
Define Homeostasis, a function of the immune system
Turn off immune responses to prevent over-reaction - remove senescent cells (for ex. red cells)
Define Surveillance, a function of the immune system
Seek out and destroy altered self (for ex. malignant cells)
T/F: The immune system can cause disease
TRUE.

It can act as barrier to stem cell therapy, transplantation and gene therapy

Innapropriate responses cause disease (immune-related inflammatory diseases)
Which of the following are immune-related diseases?

Allergy, Autoimmunity, Inflammatory bowel disease, Type 1 Diabetes, Celiac Disease, Transplantation Rejection
ALL
Define what is meant by opportunistic pathogen
A microbe that is a part of our normal flora that causes disease
List the major categories of pathogens
Viruses
Fungi
Bacteria
Protozoan Parasites
Helminth Parasites
T/F: Bacteria are eukaryotes
FALSE, they are prokaryotes, son.
What are the to shapes bacteria can take?
Bacilli (rods)

Cocci (Spherical)
What does it mean for a bacteria to be Gram +?
Its has peptidoglycan on membrane surface
What doe sit mean for a bacteria to be Gram -?
It has Lipopolysaccharide and Peptidoglycan on membrane surface
Viruses, obligate intracellular parasites, can be made from DNA OR RNA
YES
What are the shapes of viral capsids?
Icosahedral or Helical, with or without a lipid envelope
Define Protozoa
single-celled eukaryotes, transmitted by insect vectors
What are Helminths?
Parasitic worms that are highly differentiated multi-cellular eukaryotes with complex life cycles
T/F: Fungi are eukaryotes
TRUE
What are the shapes of fungi?
Round yeast cells
Filamentous Hyphae
What are the major contents of fungi cell walls and membranes?
Cell Wall: Chitin

Cell Membrane: Ergosterol
What is the main way the immune system is able to defend against things like fungi, helminths, microbes, bacteria, and protozoa?
Its ability to differentiate between self and non-self, because microbes have molecules that are different from ours (= called Pathogen-Associated Molecular Patterns, PAMPs)
Which of the following are considered "non-self" to the immune system? Flagella, Cancer Cells, Lymphocytes, Virus-infected cells, Senescent cells, Transplanted/mismatched cells, yeast, Muscle cells
Everything on list except Lymphocytes and Muscle Cells
Describe the differences between a Gram + cell wall and a Gram - cell wall
Gram +: cell wall contains ONLY Peptidoglycan, a PAMP (pathogen-associated molecular pattern), on top of a plasma membrane

Gram -: Contains Peptidoglycan, then an outer cell membrane, then Lipopolysaccharide (also a PAMP)
What are the two "arms" of the immune response?
Innate Immunity

Adaptive Immunity

Both have ways to differentiate between self and non-self
T/F: Innate immunity, the first response to a pathogen, is usually sufficient to resolve the majority of infections (i.e. no perceptible disease occurs)
TRUE
What are the characteristics of Innate Immunity?
First response to a pathogen

Usually sufficient to resolve the majority of infections

Fast, always present

Non-specific (=can recognize common elements of bacteria, viruses etc. like PAMPs)

No Memory (=won't recognize pathogen if it sees it a second time= when it sees it again it responds the same as the first time)

Limited types of responses that are non-specific for a given microbe (for ex, it recognizes peptidoglycan of all Gram+ bacteria)
T/F: Innate immunity stimulates the development of adaptive immunity
TRUE
Describe the characteristics of Adaptive immunity
Set in motion by innate immunity

Second response to a pathogen

Slower response than innate immunity b/c its tailored/adapted to deal with specific features of pathogens

Has a Memory (when it sees pathogen a second time, it remembers it and mounts a faster, stronger and more specific response)
T/F: The Recognition mechanisms for Innate immunity are variable.
FALSE. The recognition mechanisms of innate immunity are Fixed, i.e. the same response occurs every time it comes across pathogen (Adaptive immunity is variable.)
Whats the difference in time frames for Innate and Adaptive Immunity?
Innate = hours (fast)

Adaptive = days to weeks(slow)
T/F: The innate response is constant during the duration of the response
TRUE
T/F: Inflammation is a part of the Adaptive immune response
FALSE. Inflammation is a part of the INNATE immune response
What are the general steps involved in an Adaptive Immune Response?
Pathogen (say bacteria) enters via skin

Dendritic cell travels via lymphatics to Lymph node where it stimulates B cells = Abs against bacteria

B cells travel in blood back to site of infection
T/F: Neutrophils and Macrophages are only involved in Innate immunity
FALSE. They are involved in both innate and adaptive immunity
What component of the immune system causes an initial rash upon infection with a pathogen?
Innate
When a pathogen infects a host, what component of the immune response disallows it from causing more severe illness than what it initially causes?
Adaptive Immune Response

If the immune system fails to eliminate pathogen, the disease progresses to a more chronic form
T cells and B cells are what types of cells?
Lymphocytes
List some common phagocytes
Monocytes, Macrophages, Dendritic cells, Neutrophils
List some common mediator cells
Basophils, Eosinophils, Natural Killer cells, Mast cells
Where do B cells develop?
Bone Marrow (B=Bone Marrow)

Naive B cells capture antigen and become plasma cells that make antibiotics
Where do T cells, which coordinate the entire immune response, develop?
Thymus (T=Thymus)
Naive T cells are activated by what?
Antigen-presenting cells, then they become Effector T cells (because they exert an Effect!)
T/F: T cells can help B cells become what they need to be by secreting cytokines
TRUE

T cells help B cells undergo clonal expansion and differentiation into Ab-secreting plasma cells
T/F: Helper T cells (Th) are CD4+ and help B cells do their job
TRUE (T-Helper cell = 4 syllables = CD4+)
Define cytokines
Produced by T Helper cells.

They are proteins that affect the behavior of other cells
What are some of the things T Helper cells do?
Help B cells become plasma cells

Enhance Macrophage function
T/F: Interleukin is a type of cytokine
TRUE
Define Interleukin
A generic name for a cytokine produced by leukocytes

("cytokine" is a more general term)
Define Cytotoxic, and list a cell that is cytotoxic
The killing of host cells infected with intracellular pathogens (for ex. viruses)

Cytotoxic T Lymphocytes (CTL or Tc)
T/F: CTLs, or Cytotoxic T Lymphocytes, are CD8+
TRUE (Cy-to-tox-ic-T-lymph-o-cyte = 8 syllables)

(vs. T Helper cells, which are CD4+)
What cells are the effectors of Adaptive Immunity?
B and T cells
What are the two different types of Adaptive Immunity?
Humoral and Cellular
Define Humoral Immunity, a type of Adaptive immunity
Mediated by B cells and Antibodies, effective against extracellular microbes
Define Cellular Immunity, a type of Adaptive Immunity (AKA cell-mediated immunity)
Mediated by T cells

Effective against intracellular pathogens, including microbes that are ingested by macrophages and viruses that infect cells)

Cellular Immunity = T cells = Intracellular Pathogens (C-T's look INside your body = INtracellular pathogens)
Define Antigen
Any molecule that can bind specifically to an Ab or generate peptide fragments that can be recognized by the T cell's receptor
What do phagocytes do?
Seek out/take up microbes or microbial antigens by a process termed phagocytosis (hence the cell name)
T/F: Phagocytes can function as antigen-presenting cells
TRUE

Capture Antigens and present them to T Lymphocytes in lymph node
What are the 4 classes of phagocytes?
"MMDN"

Macrophages
Monocytes (take things up and KILL!)
Dendritic Cells(most potent antigen-presenting cell)
Neutrophils (take things up and KILL!)
Where do Macrophages and Dendritic cells go after they take up antigen?
Lymph Node
What do Dendritic cells do?
Circulate in tissue/blood and capture microbes and antigens then go to lymph node
T/F: Neutrophils (=Polymorphonuclear Leukocytes, or PMNs) present antigen in lymph node
FALSE.

They engulf/kill extracellular pathogens. They are NOT antigen presenting cells
List the mediator cells
"BEN M."

Basophil
Eosinophil
Natural Killer Cells (NK Cells)

Mast Cells
What do Basophils, Eosinophils and Mast Cells do?
Granulocytes that contribute to allergic responses and play a role in eliminating parasites
What do Natural Killer Cells (NK Cells) do?
Kill Virus-infected cells AND certain tumor cells
What of the following cells participate in the ADAPTIVE immune response?
B Cells
Eosinophils
T Cells
Macrophages
Mast cells
Neutrophils
Dendritic Cells
B Cells
T Cells
Macrophages
Dendritic Cells
Which of the following are antigen-presenting cells?
T Cells
Eosinophils
Dendritic cells
Macrophages
Neutrophils
NK Cells
Dendritic cells
Macrophages
Where do immune cells live?
Lymphoid Tissue

Primary: Bone Marrow and Thymus

Secondary: Tonsils, Spleen, Scattered Lymph Nodes, Mucosa-Associated Lymph Tissue
T/F: Lymphocytes stay in the lymph and only exit when called to do so by immune system
FALSE. Lymphocytes circulate continuously between blood and lymph

Cells, microbes and antigens can enter lymph nodes from the blood OR the lymph
Define Germinal Center
Site of INTENSE B Cell Proliferation (where B cells are activated)
Where is adaptive immunity induced?
Lymph Node
Where are B cells located in Lymph Node? T cells?
B Cells = Lymphoid follicle (outer edge)

T Cells = Right inside lymphoid follicle

("B Cells are on the Barrier")
Where are T cells located in the Thymus?
Cortex

T cells move towards the medulla as they mature
What percentage of T cells fail to develop or display self-reactivity, and thus are programmed to die (=called central tolerance)?
95%. This is called central tolerance
Where do Scenescent RBCs go to die?
Red pulp of spleen, which filters blood

("RBC's go to Red pulp to die")
What is contained in the white pulp of the spleen?
B cells and T cells (Spleen has aggregations of lymphocytes similar to those in lymph nodes)
How do cells and microbes enter and leave the spleen?
Blood, as opposed to using blood AND lymph like in lymph system (makes sense, since spleen filters blood, and is a secondary lymphoid organ where adaptive immune responses are initiated!)
What constitutes our normal "flora" (AKA commensal flora)
Microorganisms (bacteria+fungi)
Define Pseudomembraneous Colitis
Infection of Large Intestine with over-growth of bacteria
T/F: Some of our normal flora in gut produce antimicrobials that inhibit the growth of pathogens
TRUE
List some of the barriers in the intestine to infection
bacterial flora, mucus, glycocalyx, M cell, Paneth cells (secrete defensins), Innate immune cells in Peyer's Patches, Epithelial cells (with tight junctions), Glycocalyx (-thick sugar layer)
What are Defensins?
Anti-microbial peptides that penetrate microbial membranes, disrupting membrane

Produced by Paneth Cells (and neutrophils, epi cells)
Where do Defensins work in the body?
Intestine, Placenta, Stomach, Eyes, Oral Cavity, Urogenital Tract
What protects the stem cells of the crypts in the gut?
Paneth Cells
T/F: Receptors of the innate immune response don't change
TRUE
What are the outcomes of the innate immune response?
Inflammation (via cytokine and chemokine production)

Phagocytosis (via macrophages/neutrophils)

Cell-mediated Apoptosis (NK Cells)

Initiation of Adaptive Immunity
List some major PAMPs of different organisms
Viruses: ssRNA, dsRNA, unmethylated DNA

Bacteria: Peptidoglycan, Glycolipids, Lipoproteins, Flagellin, Inmethylated CpG Nucleotides

Gram+ Bacteria: Lipoteichoic Acid

Gram- Bacteria: Lipopolysaccharides

Fungi: Zymosan

Protozoa: Glycophosphatidylinositol
What are Toll-Like Receptors, and what do they recognize?
TLR3: detects dsRNA (viruses)
TLR4: Lipopolysaccharides (Gram- bacteria)
TLR5: Flagellin (motile bacteria)
TLR7: ssRNA (viruses)
TLR8: ssRNA (viruses)
TLR9: Unmethylated CpG DNA (Bacteria)

Viruses = TLR 3, 7, 8
Bacteria = TLR 4, 5, 9
T/F: PAMPs are not expressed by our tissues or cells
TRUE
T/F: PAMPs are not exclusive to pathogenic microorganisms but are also part of our commensal organisms
TRUE
When a TLR recognizes a PAMP, whats its signaling mechanism?
NF kappa B (an important transcription factor) = Drives inflammatory cytokine production

TLR recognizes bacteria - MyD88 - NF kappa B/Ikappa B - Pro-inflammatory Cytokines produced
What is the function of Myeloperoxidases?
Catalyze the conversion of H2O2 to Hypohalous Acid = Amplifies toxicity of ROS generated during respiratory burst
Neutrophils require what to kill bacertia and fungi?
Respiratory burst

NADPH Oxidase converts NADPH +O2 to NADPH + O2 + H=
What does Superoxide Dismutase do?
Converts H+ and O2 to Hydrogen Peroxide (H2O2) and O2.

Reverse rxm catalyzed by catalase
Define Chronic Garnulomatous Disease
Defect in NADPH Oxidase System

Immunodeficciency caused by defects in NADPH Oxidase System of enzymes that generate the superoxide radical involved in bacterial killing
Persistent bacterial infections are commonly associated with what cellular defect?
Phagocytic cellular defects
Leukocyte Adhesion Deficiency causes what?
Widespread Pyogenic Bacterial Infection
Chronic Granulomatous Disease causes what?
Intracellular and Extracellular Infection, Granulomas
G6PD (Glucose-6-Phosphate) Deficiency causes what?
Defective Respiratory Burst, Chronic Infection
Myeloperoxidase Deficiency causes what?
Defective Intracellular Killing, Chronic Infection
Chediak-Higashi Syndrome causes what?
Intracellular and Extracellular Infection, Granulomas
Whats the role of Myeloperoxidase?
Converts H2O2 to Hypohalvous Acid
What is required for Innate Immune cell homing to sites of infection?
1. Innate immune cell interaction with vascular endothelium = allows cell to leave bloodstream to enter tissue

2. Chemokines (chemotactic cytokines) = Induce directed migration of cells to sites of inflammation/infection
What molecules direct Neutrophils to sites of infection
Adhesion Molecules

Selectin-mediated adhesion is weak, and allows the neutrophils to roll along the endothelial walls of vessels
List the steps involved in neutrophil extravasation (=neutrophil leaving blood vessel)
1. Rolling Adhesion (via Selectin binding to glycoproteins)
2. Tight Binding (via LFA-1, which neutrophil up-regulates. ICAM-1, Intercellular Adhesion Molecule-1, helps too)
3. Diapedesis ("Ped" = Foot. Neutrophil makes a foot to squeexe through membrane)
4. Migration
T/F: Chemokine CXCL8 recruits Neutrophils to sites of infection
TRUE
How do NK cells (Natural Killer Cells) Distinguish between healthy and unhealthy cells?
They have an Inhibitory Receptor and an Activating Receptor

Inhibitory Receptor Recognizes "self" = prevents NK killing of host cell

Activating receptor recognizes stress signals = promotes NK killing of target cell
How do NK cells recognize "self" so as not to kill host cell?
Inhibitory Receptor recognized MHC class I, which is expressed by ALL nucleated cells

MHC1 sends neg. signal to NK cell not to kill
T/F: Natural Killer Cell Activating Receptors can detect stressed cells
TRUE

Stressed cells express unique cell surface proteins that are detected by activating receptors
T/F: Stressed host cells of a virus will express MHC class 1 chain-related protein A (MICA)
TRUE
What do virus-infected cells express that Natural Killer cells recognize that Non-infected, healthy cells do not express?
MIC Ligands for NKG2D
Define the Complement System
A group of blood and cell surface proteins that are important to inflammatory and immune responses
What are the major biological roles of the complement system?
Label Microbial Pathogens for Destruction

Disrupt Membranes of Microbial Pathogens leading to lysis

Recruit Inflammatory Cells

Remove Immune Complexes (i.e when Ab's bind antigen, they have to be removed somehow from circulation)
Which protein involved in the complement system is most important in the compliment cascade?
C3

Found in the highest concentration of all the complement proteins in the blood
What 3 pathways lead to the activation of the complement system?
Alternative Pathway = 1st to act

Lectin Pathway = 2nd

Classical Pathway = 3rd

Result of Complement Activation - Cleavage of C3 to C3a and C3b. C3b covalently bound to surface components of pathogen
Once activated, what specific events occur during the Compliment System?
Cleavage of C3 to C3a and C3b. C3b covalently bound to surface components of pathogen
Compliment system activation leads what events?
Recruitment of inflammatory cells

Opsonization of pathogens, Facilitated uptake and killing of by phagocytes

Perforation of pathogen cell membrane

ULTIMATELY: Death of Pathogen
Describe the events of compliment activation via the alternative pathway
C3 Protein floating in plasma

Small amt. C3 hydrolyzed (if this happens near pathogen, C3 is split into C3a and C3b

C3a = Inflammatory Mediator
C3b = Covalently Attaches to Glycoproteins

C3b associates with other proteins = compliment cascade continues with C5b and C5a (pro-inflammatory)

C6, C7, C8 and C9 form a complex called the MAC Comples (=Membrane Attack Complex) come in and form a hole in pathogen membrane
Where does the compliment system activity occur?
Surface of pathogen (=your cells are not affected)
Whats the most important step in compliment activation?
C3b attaching to pathogen surface
The Membrane Attack Complex is made up of what proteins in the Compliment Activation Cascade?
C5b, C6, C7, C8, C9
When does the Membrane Attack Complex form in the Compliment Activation Cascade?
After activation via any of the 3 activation pathways (Alternative, Lectin, Classical)
What protein activates Membrane Attack Complex formation by C6, C7, C8, C9?
C5b
What is Mannose' role in Compliment Activation by Lectin Pathway?
Mannose-binding lectin binds glycoproteins/lipids w/ mannose (= not common to human cells but common in bacterial cells)

=Proteases activated that produce C3 convertase which makes more C3a and C3b
List the Acute Phase Proteins
C-reactive Protein (CRP)
Mannose-binding Lectin (MBL)
Serum Amyloid Proteins (SAP)
Compliment System Proteins
Fibrinogen
What is Opsonization? What happens when we use two or more Opsonins?
Opsonization = When antigens are bound.

When we use two or more Opsonins, we get very enhanced binding and phagocytosis
What are the ways the Compliment System is Regulated?
It has to be activated

Generation and Inactivation of C3b

Compliment receptors on cells

C1 Inhibitor

Decay Accelerating Factor (DAF)

CD59 (=on cells. Protects against compliment activation)
Complement System activation is associated with what Auto-immune and Immune-mediated Diseases?
Inflammation
Vasculitis
Glomerulonephritis (renal disease)
Intra/Extravascular Hemolysis
What are the consequences of deficient production of C1 inhibitor?
Deficient C1 Inhibitor (= Inhibits C1 Activation) = We get Increased Compliment Activation = More Susceptible to infections
A lack of which protein means we can't form the Membrane Attack Complex?
C3 (We can still form the MAC complex without C2)
What clinical disease is expected in a patient deficient in compliment proteins CD59 and DAF?
Paroxysmal Noctural Hemoglobinuria = dark urine in morning bc complement activated on eurythrocytes = lysis = show up in urine
What is the most positively charged human immunoglobulin in the body?
Albumin, but it has different forms, like gamma, beta, alpha2 and alpha 1 that allow it to be more negatively charged.
List the relative charge of each of the following immunoglobulins: IgG, IgA, IgM, IgD
From more Negative to Positive:

IgG, IgA, IgM, IgD
What is an immunogen?
Any substance capable of inducing an immune response
What is an antigen?
Anu substance that can serve as the target of an immune response
What are the main determinants of immunogenicity? (=the ability of a substance to provoke an immune response)
Large Size
Chemical Complexity
Solubility and Biodegradability
Forein
Define Hapten:
A substance, usually very small, that ALONE is not immunogenic (alone, it doesn't cause an immune response) but after conjugation to a carrier protein or cell becomes immunogenic. Ab formed can then bind the hapten, carrier, or both.
Define Epitope
The portion of the antigen that is bound by the Antibody or T cell. (the portion of the antigen that determines its immunogenicity)

Epitopes can be different on the same molecule or repeated on the same molecule
Whats the difference between a Linear Epitope and a Discontinuous Epitope?
Describe the response of an antibody at initial contact and then when it contacts antigen again
Memorize the following chart:
Where do the hyper-variable regions lie in the structure of an immunoglobulin?
The hyper-variable regions are in the part of the immunoglobulin available for contact with antigen
Define Isotype
Variants present in all members of the species
Define Allotype
Variants that differ between members of the species
Define Idiotype
Variants due to the hypervariable regions of Ig and TCR
What are some of the forces that act to bring Ab into contact with Antigen?
Hydrogen bonding
Electrostatic
Van Der Waals
Hydrophobic
What are the events leading up to antibody secretion by a plasma cell?
Antigen recognition induces expression of effector molecules by T cell which activates B cell

B-cell proliferation

Differentiation to resting memory cells and Ab-secreting plasma cells
How are antibodies used in clinical medicine?
1. Auto-reactive Ab in autoimmune diease
2. Vaccination and use of pre-formed Ab
3. Monoclonal Ab
4. Laboratory Serology
Describe Immunoflourescence
Produce reagents specific for what we're measuring in a serum sample, for ex, use an Anti IgA = an Ab specific for IgA

The dye is labeled and allows you to video it.
Whats the idea behind vaccination?
Give pt. vaccine --> produces a primary immune response --> Lots of memory cells produced --> when person encounters pathogen in real life, the response to pathogen will be quicker bc memory cells already present (primary IgG Abs will be present now, which are a better isotype for the immune response because they are More specific than monoclonal Abs)
How can we mass produce monoclonal Abs for market?
Infect an animal with antigen

Remove spleen, which should have lots of B cells proliferating

Spleen will produce monoclonal Abs, however these Abs are usually different that the human form of the same Ab, so thay have to be altered before they will work in humans
Define Serology
Lab detection of specific serum Abs

For ex. ELISAs
What are ELISAs?
Enzyme-linked immunosrobent assay

Used to detect an semi-quantitate antigen or Ab

Reagent Ab labeled with detector so we can quantitate it.
A pt. presents with the following laboratory values. What is your initial diagnosis?

IgA = 5 mg/dL (Normal = 40-60)

IgG = 1320 (Normal = 800-1400)

IgM = 72 (Normal = 30-120)
Pt. has immune deficiency with selective IgA deficiency (this is the most common immune deficiency)
A pt. is having sinusitis, continuous cold, and nasal blockage after having moved to a new city. If his Immunoglobulin Levels are as follows, whats your initial diagnosis?

IgA = 120 (N=76-390)
IgE = 1500 (N=0-380)
IgG = 1280 (N=650-1500)
IgM = 300 (N=40-345)
Patient's IgE is high, meaning he may be experiencing an allergic response since IgE is involved in sensitization of mast cells
List the Immunoglobulin proteins in order from lowest serum concentration to highest.
"Ed Did Great Getting Mary Awake 2 GoGo"

(Just remember the G's go in order from lowest conc. = G4 to G3 to G2 to G1 = highest conc.)

IgE -- IgD -- IgG4 -- IgG3 -- IgM -- IgA -- IgG2 -- IgG1
What is the role of the Fc Receptor?
Complement System Activation

Neutrophils etc. will have them.
Whats the difference between secretory IgA and Serum IgA?
Secretory IgA = main Immunoglobulin found in mucous secretions like nasal, tears, saliva

Serum IgA = in serum
Compliment System Events
What is the signaling complex of the T Cell? (i.e. what part of the T Cell reads whether a cell is an antigen or not?)
CD3
Define T Cell
Primary initiator, coordinator, regulator of Adaptive Immune response.

Both cellular and humoral immunity depend on T cell Function
What are the three major functions of T cells?
"Help, Kill, Regulate"

1. Helper Cell - help B cells make Abs. Secrete cytokines that recruit immune cells and activate immune and non-immune cells

2. Killer Cells - Mediate direct apoptosis of virus-infected cells and tumor cells

3. Regulatory Cell - prevent immunopathology by suppressing immune responses
Where do T cells originate, and where do they migrate to?
Originate in Bone Marrow, migrate to Thymus where they undergo selection

Become active in Lymphoid organ after they encounter antigen

Once activated, T Cells go to site of infection and perform their effector function
Describe the T Cell Receptor
Antigen Recognition occurs at alpha and beta sites

CD3 complex on either side of alpha/beta read whether T cell is seeing an antigen

Disulfide bonds attach the signaling proteins to the receptor
T/F: The T Cell Receptor (TCR) only recognizes antigen presented by MHC (=Major Histocompatibility Complex)?
TRUE

T Cell Receptors can either be CD4+ or CD8+

CD4+ binds MHC class II

CD8+ binds MCH class I

To remember: CD4 x 2 = 8
CD8 x 1 = 8
In what places throughout the body can the T Cell Receptors recognize MHC Class I and MHC Class II molecules?
Thymus (As T cell progenitors)

Secondary Lymphoid Organs (as mature Naive T Cells)

Periphery (skin, intestine, lungs, etc. as activated T cells)
What percentage of T Cells Fail to become selected and activated in the thymus, and are thus apoptosed?
95%
T/F: Only self antigens are produced in the thymus
TRUE
T/F: The only antigen that T Cells test themselves with are other anitgens
TRUE
Where in the thymus does positive T Cell selection occur? Negative T Cell selection?
Positive selection occurs in the cortex of the thymus

Negative selection occurs in the Medulla of the thymus
How does positive T cell selection in cortex of thymus work?
T cells that recognize antigen presented by MHC survive then go on to undergo negative selection.

If they don't recognize MHC, the T cell is deleted.
Once a T cell in the thymus recognizes an antigen presented by MHC, it then goes on to negative selection - describe how negative selection works
T cells that recognize "self" peptides are deleted, because we don't want T cells attacking our own cells! This is a safety mechanism

If the T cell does not recognize the "self" peptide, it survives and leaves the thymus as a mature naive T cell.
What is the determining factor in T Cell Negative selection in the medulla of the thymus
Binding affinity (Remember; negative selection is when T Cell is tested to see if it binds "self")

T Cell binds Moderately or Loose = Lives

T Cell binds tightly = It is Killed
Define the concept of central tolerance
Positive and Negative selection generate Central Tolerance (= occurs in thymus) vs. Peripheral Tolerance that plays a role in preventing autoimmunity
Regulatory T Cells (Tregs) do what?
Suppress Adaptive Immune Response by suppressing action of CD4 and CD8 cells
Negative selection generates a small number of what type of cell that aids in inactivating self-reactive T cells?
CD4+ Regulatory T cells (Tregs)

Negative Selection = T cells presented with "self" antigen. Those that bind "self" are killed, however a small number survive and become CD4+ regulatory T cells (Tregs) that REGULATE (!) those cells that may have escaped these regulatory mechanisms to prevent T cells from attacking self.
Tregs are generated in what two ways?
1. Natural Tregs generated via Negative Selection (mediate peripheral tolerance)

2. Induced Tregs are generated in the periphery (ALSO mediate peripheral tolerance)
T/F: Both Natural and Induced Tregs mediate peripheral tolerance
TRUE
Lack of Tregs results in what condition? What are the signs/symptoms?
A defect in peripheral tolerance and an inability to inactivate T and B cells that escape central tolerance

Signs/Symptoms: watery diarrhea, failure to thrive, dermatitis, autoimmune diabetes in infnacy

Treatment = bone marrow transplant
T/F: T Cells circulate throughout the blood and through secondary lymphoid tissue in search of Antigen presenting cells presenting something it can recognize.
TRUE
Where do T cells mature?
Blood
What types of Antigen-Presenting Cells occur in the Lymph Nodes?
1. Professional Antigen presenting cells (macrophages, dendritic cells)

2. B Cells
What types of Antigen-Presenting Cells occur in the Periphery (as opposed to the lymph node)?
1. Professional Antigen-presenting cells (same as in lymph node - macrophages, dendritic cells

2. Non-Immune cells, such as virus-infected epithelial cells
What are the four CD4+ T Helper Cell Subsets?
Th1
Th2
Treg
Th17
What is the function of Th1 cells, a subset of CD4+ T Helper cells?
Initiate/Regulate CELLULAR Immunity (vs. Th2 cells which do HUMORAL immunity)

IFN-gamma (AKA Type II Interferon) = a cytokine that helps to activate macrophages and help B cells make Ab (IgG1). In this way, Th1 cells also help with humoral immune response
What is the function of Th2 cells, a subset of CD4+ T Helper cells?
Initiate/Regulate HUMORAL immunity (vs. cellular immunity regulated by Th1 cells)

Recognize peptide presented by B Cells in lymph nodes

Secrete cytokines that help B Cells
What are two anti-inflammatory cytokines that inhibit Th1 development?
IL-10 and transforming growth factor beta (TGF-beta)
What do Th17 Cells do?
Produce pro-inflammatory cytokines (= play role in inflammatory bowel disease, Rheumatoid Arthritis, MS and Asthma)

IL-17, IL-21, IL-22 Activate macrophages, fibroblasts, endothelia and epithelia to promote inflammation
When you see Th17 Cell, what process is most likely occurring?
Inflammation
What are the two types of CD4+ T Cell Receptors?
Alpha-beta and gamma-delta = the regions that bind (vs the CD3 region of the protein)

vs. CD8+ T cells
CD8+ cells recognize antigen presented in the context of MHC I or MHC II?
MHC I
Note the different roles of CD4 and CD8 Cells
List the different CD4+ Effector T cells, the Cytokines produced by each CD4+ T Cell effector, and their cellular target.
CD4+ Effector T Cells Include: Th1, Th2, Th17, Treg

NOTE: There is an error on picture; Treg effector T cells produce IL-10 and TGF-beta, NOT IL-1.
CD4+ T Cell Effector Th1:

Cytokines Produced?
Cellular Target?
Cytokines Produced =

IL-2
IFN-gamma
Cellular Target =

Macrophages
CD8 T Cells



CD4+ T Cell Effector Th2:

Cytokines Produced?
Cellular Target?
Cytokines Produced =

IL-4, 5, 6, 10, 13
Cellular Target =

B Cells
Mast Cells
Eosinophils
CD4+ T Cell Effector Th17:

Cytokines Produced?
Cellular Target?
Cytokines Produced =

IL-17, 21, 22
Cellular Target =

Fibroblasts
Endothelial Cells
Epi cells
CD4+ T Cell Effector Treg:

Cytokines Produced?
Cellular Target?
Cytokines Produced =

IL-10, TGF-beta
Cellular Target =

Th1, Th2, Th17
T Cell Cytokine IL-2:

Major Source?
Major Effect?
Major Source =

CD4+ cells
Major Effects =

Proliferation of T, B, NK cells
T Cell Cytokine IL-4:

Major Source?
Major Effect?
Major Source =

Th2 T Cells
Major Effects =

B-cell Proliferation
IgE Production
Induces Th2 Cell development
T Cell Cytokine IL-10:

Major Source?
Major Effect?
Major Source =

Th2 T Cells
Treg
Major Effects =

Inhibits macrophages and dendritic cells
Inhibits development of Th1 cells
T Cell Cytokine IL-12:

Major Source?
Major Effect?
Major Source =

Th1 T Cells
Major Effects =

Inhibits development of Th2 cells
Promotes development of Th1 cells
T Cell Cytokine IL-13:

Major Source?
Major Effect?
Major Source =

Th2 Cells
Basophils
Mast Cells
Major Effects:

IgE Production
Increased Mucous Production


T Cell Cytokine IL-17:

Major Source?
Major Effect?
Major Source =

Th17
Major Effects =

Induce inflammatory cytokines and recruit neutrophils
T Cell Cytokine INF-gamma:

Major Source?
Major Effect?
Major Source =

Th1 Cells
Major Effects =

Macrophage Activation
Promotes Th1 Development
Promotes MHC Expression
T Cell Cytokine TGF-beta:

Major Source?
Major Effect?
Major Source =

Treg
Major Effects =

Inhibits Lymphocyte Activation
Anti-inflammatory
Promotes IgA Production
MHC Class I is composed of:
A. alpha chain non-covalently associated with B2-microglobulin
B. alpha chain covalently associated with B2 microglobulin
C. Covalently associated alpha and beta chains
D. non-covalently associated beta chains
A. MHC Class I is composed of:

Alpha chain non-covalently associated with B2-microglobulin
What are Exogenous antigens loaded onto?
MHC Class II molecules in an endosome
Why is tissue transplant difficult from patient to patient?
MHC molecules are very different = why tissue transplant is difficult
What is the major histocompatibility complex?
The genetic region that contains the MHC I and MHC II genes
What determines compatibility between individuals during tissue and organ transplantation?
MHC I and MHC II determine Histocompatibility
What do MHC molecules do?
Present peptides to T cells
Describe the relative size of epitope MHC Class I and MHC class II molecules can bind
MHC I binds peptides (epitopes) 7-15 AAs in length, while MHC II binds peptides (epitopes) 10-30 AAs in length
Picture the MHC class I and MHC class II molecules
Antigens present outside the cell are presented to CD4+ T cells by what? (Exogenous antigens taken into cell, degraded, presented)
MHC II
Antigens present inside the cell are presented to CD8+ T cells by what? (Endogenous antigens in cytosol degraded, presented)
MCH I
Antigens outside cell are presented on which MHC?
MHC II
Antigens inside cell are presented on which MHC?
MHC I
How do antigen-presenting cells take in antigen?
Receptor-mediated endocytosis:

-Antigen coated with complement proteins --> Antigen taken in by complement receptor

-Fc Receptor

Antigens can ALSO get in by fluid-phase endocytosis
List the pathways dendritic cells use to get antigens into them
1. Receptor-mediated endocytosis (MHC II = CD4+)
2. Macropinocytosis (MHC II = CD4+)
3. Viral Infection (MHC I = CD8+))
4. Cross-presentation after phagocytic or macropinocytic uptake (MHC I)
5. Transfer from incoming dendritic cell to resident dendritic cell (MHC I)
Where are peptides that bind MHC Class II Molecules generated?
Acidified Endocytic Vesicles
Acidification of vesicles does what to proteases which degrade antigen into peptide fragments?
Acidification of vesicles ACTIVATES proteases to degrade antigen into peptide fragments

Vesicles containing peptides then fuse with vesicles containing MHC Class II molecules
Peptides that bind MHC II molecules are generated in what?
Acidified Endocytic Vesicles
T/F: Acidification of vesicles activates proteases to degrade antigen into peptide fragments
TRUE
Where does processing of antigens for presentation on MHC II occur?
Phagolysosome
Picture the process of antigen presentation on MHC II
T/F: Viruses use the cell's innate machinery to replicate, thus viral proteins are synthesized in the ER and golgi, and assembled in the cytosol
TRUE

Viruses are obligate intracellular parasites
Is a protein transporter required to get protein peptides into the ER?
Yes,

Proteins required for peptide transport into the ER for loading into MHC I molecules
Where does processing of antigens for presentation on MHC I occur?
ER

(Vs. for MHC class II, which occurs in phagolysosome)
T/F: An MHC molecule can present a variety of peptides
TRUE

Different from Ab's, which can only bind one thing = Ab's more specific than MHC's
T/F: A T Cell receptor can bind a variety of peptides presented by an MHC molecule
FALSE

B and T Cell receptors stick to one peptide
What types of cells express MHC Type I?
T cells
B Cells
Macrophages
Dendritic Cells
Neutrophils

ALSO:
Thymic Epithelium
Liver Hepatocytes
Kidney Epi
Brain
What Cells Express MHC Class II?
T Cells, when activated
B Cells
Macrophages
Dendritic Cells
Thymic Epithelium

NOT NEUTROPHILS, Liver hepatocytes, Kidney Epithelium, Brain
What is Bare Lymphocyte Syndrome?
A disease in which EITHER MCH class I or II molecules NOT expressed on cell = Immunodeficiency

Deficiency/Lack of MHC Class I = Lack of CD8+ T cells = Susceptible to intracellular pathogens

Lack of MHC Class II = Lack of CD4+ = susceptible to extracellular pathogens
Define Human Leukocyte Antigen (HLA)
Genetic designation for human MHC

For ex. HLA-A*0201

HLA = Loci, 0201 = Alleles = we want to match as many of these between donor/recipient as possible
What are major histocompatibility complex molecules?
Highly Polymorphic Proteins that bind antigens and present them to T cells (HMC I and MHC II)
Define Polymorphism
Existence of different variants of a gene; 2 or more different forms or alleles of a gene
Define Allele
Natural variants of a gene; one of several alternate forms of a gene at a single locus (HLA-A has 506 alleles etc.)
Define Haplotype
The set of polymorphic genes carried on a single chromosome. Humans inherit two haplotypes, one from each parents.
T/F: MHC isotypes differ in the extent of their polymorphism
TRUE

Polymorphism can mean you are expressing two forms of one gene
Which MHC isotypes encode MHC Class I?
HLA-A
HLA-B
HLA-C

(Not as important to know: HLA-E, HLA-F, HLA-G)
Which MHC isotypes encode MHC Class II?
HLA-DP
HLA-DQ
HLA-DR

(Not as important to know: HLA-DM, HLA-DO)
What characteristic of HLA genes has important implications for tissue transplation?
The fact that some HLA genes are highly polymorphic = there are different variants of the gene
HLA-A Encodes what?
MHC class I alpha chain

(Beta chain doesn't really contribute much - just there for structural support)
HLA-DR Encodes what?
HCM Class II alpha and beta chains

(unlike the important HLA-A, HLA-B, and HLA-C mentioned in class, which only encode alpha chain)
T/F: Expression of MHC alleles is co-dominant
TRUE

Results in heterozygosity - inherit dif. alleles from both parents = both alleles expressed
Why is the fact that Expression of MHC alleles is co-dominant = heterozygosity good for a person?
Good b/c this means we have immunity to many dif. pathogens.

If we were homozygous, we wouldn't be as immune to as many things.
T/F: All MHC molecules can be expressed on the cell surface at any given time
TRUE

All of them can bind and present a range of peptides
Besides polymorphism, what else contributes to the diversity of MHC molecules expressed by an individual?
Polygeny
Which regions of the MHC I molecule are the most diverse in terms of amino acid variability?
alpha 1 and alpha 2 domain = most diverse
Which regions of the MHC II molecule are the most diverse in terms of amino acid variability?
alpha 1 and beta I regions = most diverse
Where is variation between MHC allotypes concentrated in MHC I and MHC II molecules?
Variation occurs at the sites that bind peptide and the TCR
Whats one reason some people survive diseases that others don't?
Diversity of MHCs in the person
T/F: MHC heterozygosity has no affect in the progression of AIDS in HIV-1 infected people
FALSE.

MHC Heterozygosity delays the progression of AIDS in HIV-1 infected people

Inc. Heterozygosity = Longer Survival
What cell confers antigen specificity?
Lymphocytes
Where do WBCs undergo hematopoesis?
Bone Marrow
Whats the difference between B cells and T cells in their maturation processes?
T cells leave Thymus to mature and become functional T cells

B cell precursors remain in Bone Marrow to mature (from pluripotent stem cells)
Describe how Lymphocytes confer antigen specificity
1. Cells give rise to lots of lymphocytes that circulate - each has a dif. cell surface receptor
2. Receptors of only a few lymphocytes interact with any given pathogen
3. Pathogen-reactive lymphocytes triggered to divide/proliferate
4. Pathogen-activated lymphocytes differentiate into effector cells that eliminate pathogen
Describe the structure of Immunoglobulin
2 Identical light chains

2 Identical heavy chains

These two things make up the antigen binding site = where the variability occurs
Variable regions of immunoglobulin are encoded by what?
gene segments:

2 gene segments code for te 2 light chains

3 gene segments code for the 2 heavy chains

SO cells, as they develop, have many of these gene segments to choose from
How does a B Cell produce a Kappa Light Chain?
The variable part of the LIGHT chain is encoded by two dif. gene segments: V Kappa and J Kappa.

V Kappa = 40 to choose from
J Kappa = 5 to choose from

B Cell chooses one of each to form the coding message
What genes help out in the formation of B Cell Kappa Light Chain Rearrangement?
RAG-1 and RAG-2

Recombination Activating Genes 1 and 2
What do RAG-1 and RAG-2 do?
Encode proteins/ enzymes involved in function of choosing gene segments, splicing them together, taking out everything else not needed.
The B cell is teh only cell to do what in terms of immunoglobulins?
B Cell is the only cell that can Re-arrange gene segments, which is why the B cell is the only one to make Antibody
What determines the isotype of the Immunoglobulin?
Constant region of the heavy chain
What are one of the 1st antibodies produced by B cells?
IgM
The heavy chain is encoded by how many VARIABLE segments?
3

V, D, J

(where as Light chain has 2 variable segments, V and J)
How is Antibody diversity created?
Developing B cell receptors form from gene segments

Variable regions encoded by V, D, J segments

Random combination of segments produces greater variability
What are additional ways Antibody diversity can be created?
Differences in the ways genes come together

Terminal Deoxynucleotidyl transferase (TdT)

Light and Heavy chain combinations

Somatic Hypermutations
How does Terminal Deoxynucleotidyl Transferase (TdT) produce increased Antibody diversity?
TdT = An Enzyme active in developing Lymphocytes

Adds random nucleotides to segments
Whats the difference between B cells and T cells diversity in terms of affinity for antigens?
B cells can undergo somatic hypermutations = point mutations increase their diversity for antigens they recognize = dif. antigen recognized

After being cloned, T cells still have the SAME specificity for antigen
How do Somatic Hypermutations produce increased Antibody diversity?
Point mutations occur in receptor molecules = changes affinity for receptor
What are the surface Immunoglobulin receptors?
IgM or IgD
Which Immunoglobulin receptors do intracellular signaling?
IgA (Ig-alpha) and IgB (Ig-beta)

-Can be phosphrylated and then do intracellular signaling

Accessory proteins = mechanism of action for signal being carried from membrane to nucleus = to divide, produce cytokines etc.
What Co-stimulatory molecules are involved in B-cell Activation?
CD21 and CD19

Part of the co-receptor complex
What are the 5 major Antibody Isotypes?
IgM
IgD
IgC
IgE
IgA
If a cell is making IgM, hwo do we get production of the other isotypes?
When B cell activated, many of the clones produced will undergo isotype switching = switch from making IgM to making some other isotype
Isotype switching does what to the VDJ segments?
Brings them closer
Both somatic hypermutation AND isotype switching are dependent on what?
AID, Activation-Induced Cytidine Deaminase
AID, Activation-Induced Cytidine Deaminase is expressed only in what?
ACTIVATED B Cells
How does AID, Activation-Induced Cytidine Deaminase work?
Makes Knick in DNA = DNA Degraded = VDJ Segments for Variable Region coem close = Everything between them is gone = Now daughter cell will make a dif. Ig
What influence do T Cells have on isotype switching? (i.e. B cells making a dif. Ig?
T Cells produce cytokines that influence isotype switching (for ex. interleukin 4 = induces switching to IgE = Why TH2 cells imp. in allergic reactions)
What determines what isotype is produced when a B Cell undergoes isotype switching?
Helper T Cells + B Cells = Activation of B cell so it can do isotype switching

CD40 on B Cell
AND
CD 40 on T Cell
Whats the major function of IgM?
Activation of Complement System
Whats the major function of IgD?
not on chart
Whats the major function of IgG1?
Neutralization
Opsonization
Sensitization for killing by NK cells
Activation of Complement System

NOTE:
Transported across placenta
Diffusion into Extravascular Sites
Whats the major function of IgG2?
Neutralization

Note:
Diffusion into Extravascular Sites
Whats the major function of IgG3?
Neutralization
Opsonization
Sensitization for Killing by NK cells
Activation of Complement System
Somewhat: Sensitization of Mast Cells

Note:
Transported across Placenta
Diffusion into Extravascular Space
Whats the major function of IgG4?
Neutralization

Note:
Transported across Placenta
Diffusion into Extravascular Space
Whats the major function of IgA?
Neutralization
Somewhat: Activation of complement system

Note:
Transport Across Epithelium
Diffusion into Extravascular Sites
Whats the major function of IgE?
Sensitization of Mast Cells

Note:
Diffusion into Extravascular Sites
After antigen exposure, what is the primary response made up of mostly, in terms of Immunoglobulins isotype?
IgM isotype
After antigen exposure, what is the secondary response made up of mostly, in terms of Immunoglobulins?
IgG isotype
What is Burkett's Lymphoma?
B Cell Malignancy caused by a virus = produces mono

Infects B cells, and, via chromosomal rearrangement, induces cancer
During B Cell replication, translocation from chromo 8 to chromo 14 results in what?
Chromo 14 contains genes for the heavy chain of Ab. If transolocation puts a protooncogene on chromo 14, it can be cancerous
Protection in the immune system of a developing fetus or neonate comes from what?
IgG from mother

5-6 months later, babies' IgG goes down, but by this time baby can develop own Abs.
When are babies most susceptible to infections?
After birth, before they've begun to fully make their own IgG (i.e. couple months after birth)
What happens if someone doesn't have RAG-1 or RAG-2?
Severe Immunodeficiency = don't produce enough T cells or B cells

= "Bubble Boy Syndrome"
What happens if someone doesn't have any AID, Activation-Induced Cytidine Deaminase?
No isotype switching = Lots of IgM, not a lot of others = very susceptible to infections
When dendritic cells recognize pathogen via pattern recognition, what are some of the things they do?
Undergo functional maturation

Begin to present peptide on their cell surface

Up-regulate co-stimulatory molecules
Chemokines secreted by what Cell types recruit dendritic, T cells and B cells to lymph nodes?
Lymph node stromal cells

Dendritic Cells
What is the most potent antigen presenting cell?
Dendritic Cells
What are some examples of dendritic cells on the skin? Tissues? Blood?
Skin: Langerhan's and Dermal dendritic cells

Tissues: Myeloid dendritic cells

Blood: Plasmacytoid dendritic cells
What do Langerhan's Cells (epidermal dendritic cells) preferentially induce; cellular immunity or humoral immunity?
Cellular Immunity
What do Dermal dendritic cells (epidermal dendritic cells) preferentially induce; cellular immunity or humoral immunity?
Humoral Immunity

Secrete cytokines that promote the development of Ab responses
Whats the major Type I Interferon producing cell in the blood?
Plasmacytoid Dendritic Cell

Very good at detecting viruses in blood

Make Type I Interferons (IFN-alpha, INF-beta) that play a major role in anti-viral defense
T/F: Myeloid dendritic cells are found only in lung and intestinal cells
FALSE

Myeloid dendritic cells are found in almost all tissues

If from monocytes, they are inflammatory monocytes
Dendritic Cells collect antigens and migrate where to present a now processed antigen?
Lymph Node
Immature Dendritic cells in the peripheral tissues seek out antigen, then what?
They mature as they migrate to lymph node where they present a processed antigen
Dendritic cells change their morphology and function after collecting antigens: What is the morphology of an immature dendritic cell?
MHC II co-localizes with lysosomal marker
Dendritic cells change their morphology and function after collecting antigens: What is the morphology of a dendritic cell after antigen uptake?
MHC class II is loaded with antigen and moves OUT of lysosome to cell surface = this is a major change in the membrane
What things occur during dendritic cell maturation?
1. Endocytosis is down-regulated
2. Antigens undergo processing into peptides
3. Peptides loaded onto MHC molecules
4. MHC molecules move to cell surface and display peptide
5. Dendritic cells down-regulate chemokine receptors that promote migration to inflammatory chemokines
6. Dendritic cells up-regulate chemokine receptors that promote migration to lymph node-derived chemokines
7. Dendritic cells up-regulate expression of co-stimulatory molecules (CD80 and CD86)
What is Chemotaxis?
Process of directed migration down chemical gradient

ie infection = inflammation and release of inflammatory chemokines
What chemokine receptors respond to the CCL5 chemokine?
Chemokine Receptors CCR1 and CCR5
During infection, dendritic cells leave tissues and migrate in response to what?
Lymphoid chemokines present in lymphatics and lymph nodes
What chemokine receptor respond to the CXCL12 (SDF-1, stromal-derived factor 1) chemokine?
Chemokine Receptor 4 (CXCR4)
What chemokine receptor responds to the CCL19 and CCL21 chemokines?
Chemokine Receptor 7 (CCR7)
What do inflammatory chemokines do?
Recruit phagocytes to sites of infection where they destroy pathogens and/or acquire antigens for processing and presentation to T Cells
During infection, What do professional antigen-presenting cells respond to?
Lymphoid Chemokines = directs them to lymph nodes for interaction with T cells
T/F: T cells can access lymph from blood and lymph
TRUE
Once antigen-presenting cells are in the lymph node, whats the next step?
T Cells enter lymph node, crossing High Endothelial Vacuoles, and monitor the antigens presented by macrophages and dendritic cells

If T Cells don't encounter specific antigen, they leave

T Cells that encounter specific antigen proliferate and differentiate into effector cells
What processes are needed to activate T cells once they encounter a specific antigen?
Adhesion (to antigen-presenting cell)
TCR-MHC Peptide
Co-Stimulation
Cytokine IL-2
Integrins and Adhesion molecules are composed of what?
alpha and beta chains
What is LFA-1?
Leukocyte Function Associated Antigen 1

Integrin expressed by T cells that allows binding to the adhesion molecule ICAM-1 on Antigen Presenting Cells
Picture Adhesion, with LFA-1 and ICAM-1
Without expression of CD18, what happens?
T cells can't be activated (LFA-1)

Macrophages can't get out of blood to access sites of infection (MAC-1 binds ICAM1 expressed on endothelial cells)

i.e. without CD18, MAC1 can't interact with ICAM1
What is Leukocyte Adhesion Deficiency (LAD)?
Defect in the beta subunit of MAC1
During the TCR-MHC (T Cell Receptor/MHC) Peptide process of T Cell Activation, Antigen is present via which MHC to CD4+ cells?
MHC II
During the TCR-MHC (T Cell Receptor/MHC) Peptide process of T Cell Activation, Antigen is present via which MHC to CD8+ cells?
MHC I
T/F: Antigen presented via MHC's to CD4 o4 CD8 cells is sufficient for T cell Activation
FALSE

We must co-stimulate
What does co-stimulation involve?
B7 Molecules = CD80 and CD86 Human Antigens
What do B7 molecules (CD80 and CD86) bind to?
CD28 on the T Cell
Whats the first cytokine produced after T cell activation and a major consequence of T Cell Receptor signaling?
IL-2
Activated T cells express IL-2 which does what?
IL-2 enhances T cell proliferation and differentiation in an autocrine fashion
What is MHC Restriction?
When the T Cell Receptor recognizes a complex of peptides bound to MHC = the T Cell Receptor is specific for the combination of peptide and MHC = BOTH have to fit
T/F: Antigen presentation to T Cells is MHC restricted
TRUE

alpha and beta chains can recognize antigen AND the class II molecule

BUT, Not all antigens are presented this way (for ex. Super antigens)
What do Super Antigens do?
Bridge the T Cell Receptor to MHC Receptor = Super antigen can bind = Polyclonal Activation
T/F: MHC Molecules are never on the surface unless antigen is presented
TRUE
What do B7 Molecules (CD80 and CD86) bind?
CD28 on BOTH CD4+ and CD8+ T Cells
What is Tolerance?
A way the immune system prevents autoimmunity
T/F: It is equally important to turn immune responses on and off
TRUE
B7 Co-stimulatory molecules CD80 and CD86 bind to CD28 on the T Cell to do what?
Activate T Cell
T/F: B7 Co-stimulatory molecules can also Suppress the T Cell?
TRUE
Shortly after being activated, what is Up-regulated on the T Cell?
CTLA-4 (Cytotoxic T Lymphocyte Antigen 4)
T/F: Antigen presentation to T Cells is MHC restricted
TRUE

alpha and beta chains can recognize antigen AND the class II molecule

BUT, Not all antigens are presented this way (for ex. Super antigens)
What do Super Antigens do?
Bridge the T Cell Receptor to MHC Receptor = Super antigen can bind = Polyclonal Activation
T/F: MHC Molecules are never on the surface unless antigen is presented
TRUE
What do B7 Molecules (CD80 and CD86) bind?
CD28 on BOTH CD4+ and CD8+ T Cells
What is Tolerance?
A way the immune system prevents autoimmunity
What are the two types of Tolerance?
Central and Regulatory
T/F: It is equally important to turn immune responses on and off
TRUE
B7 Co-stimulatory molecules CD80 and CD86 bind to CD28 on the T Cell to do what?
Activate T Cell
T/F: B7 Co-stimulatory molecules can also Suppress the T Cell?
TRUE
Shortly after being activated, what is Up-regulated on the T Cell?
CTLA-4 (Cytotoxic T Lymphocyte Antigen 4)
What does CTLA-4 (cytotoxic T Lymphocyte antigen 4 do?
After T cell is activated its up-regulated and LIMITS the T Cell's Proliferation

How: It has greater affinity for CD80 and CD86 on Antigen presenting cell than CD28 (which is on T cell)

OVERALL EFFECT: T Cell Proliferation limited
What do Tregs (Regulatory T Cells) do?
Mediate peripheral tolerance
What are the two types of Tregs (Regulatory T Cells)?
Natural Tregs (ie Naive T Cells) = Generated in thymus during positive and negative selection

Induced Tregs (ie Effector T Cells) = Generated in Periphery
How do Tregs (Regulatory T Cells) work?
Secrete TGF-beta
Interact with T Cell
Down-regulate/inhibit function of dendritic cell
When antigen is presented by Non-Professional Antigen presenting cells, what happens?
T Cell Tolerance
Suppression of T Cell responses occurs by which mechanisms?
Antigen presentation in the absence of co-stimulation (= we need B7 = CD80/CD86) to activate T Cell

Tregs
Differentiate between NaiveT Cell Response and Effector T Cell Response (=Th1 or Th2)
Naive T Cell's require co-stimulation (=via B7 = CD80/CD86)

Effector T Cells do NOT require co-stimulatory molecules to perform their functions
What does B Cell (=Lymphocyte) activation require?
CD4 T cell help for T-dependent antigens
Whats the process of B Cell activation?
B cell endocytoses antigen bound to membrane immunoglobulin --> processes then presents antigen to CD4 T Cell
T/F: B Cells are professional antigen-presenting Cells
TRUE

Antigen processed then presented on MHC II
What on the T Helper cell membrane and the B cell membrane is required for B cell activation and T Helper cell activation?
CD40 Ligand (T Helper cell) and CD40 (B Cell)

= Co-stimulation = now T cell knows its interacting with an antigen-presenting cell
After B Cells internalize (via receptor mediated endocytosis) and process antigen bound to B cell receptor, what happens?
Antigen is processed and presented on MHC class II
How do T cells activate B cells?
T cells secrete cytokines, like CD40L (=CD40 Ligand) which attaches to CD40 receptor on B cell
T/F: Most antigens are T-dependent (-Thymus dependent) and T cell help is required for B cells to produce antibodies
TRUE

HOWEVER-->

The exception is that B cells DO NOT need T Cell help to respond to Thymus-INDEPENDENT antigens
What are the two antigens that can activate B cells without the help of T cells?
TI-1 and TI-2

HOWEVER-->
Some cytokines can induce isotype switching in B cells in the absence of T cell help
T/F: When B cells are activated by T-independent antigens without T Cell Help by TI-1 antigens, cytokines are not produced and thus IgM antibody is the only one produced (ie no isotype switching)
TRUE
T/F: When B cells are activated by T-independent antigens without T Cell Help by TI-1 or TI-2 antigens, there is no affinity maturation (=low affinity Abs)
TRUE
When B cells are activated by T-independent antigens without T Cell Help by TI-1 or TI-2 antigens, No memory response is generated.
TRUE
What is LPS?
A TI-1 antigen (=Thymus Independent Type 1 Antigen)

Thus, with exposure to LPS, we will get Anti-LPS IgM antibody response b/c LPS is a TI-1 antogen = they induce IgM Abs only
What does LPS antigen (=a TI-1, Thymus Independent Type 1 antigen) interact with?
TLR-4 = B cell is interacted without any T Cell help = the definition of TI-1 and TI-2 antigens
T/F: LPS TI-1 antigen can induce an antibody response to other antigens
TRUE

For. ex. LPS can induce an antibody response to a flagella if it comes into contact with B cell
What are TI-2 (=Thymus-Independent Type 2 antigens) antigens composed of?
Repetitive Carbohydrate (polysaccharide) or protein epitopes
What do TI-2 antigens result in?
Extensive BCR and co-receptor cross-linking
What is the Hib vaccine (H. Influenza) composed of?
Tetanus toxoid (=a T-dependent antigen) and H. Influenzae Polysaccharide (=a Gram neg. - capsule = a T-Independent Antigen)
Why is the HiB Vaccine effective? (=H. Influenza, or Hemophilus Influenza)
Tetanus toxoid converts the polysaccharide into a T-dependent antigen
How do cytokines work?
Autocrine
Paracrine
Endocrine
What are some examples of cytokines?
Hematopoietic Growth Factors

Chemokines

Interleukins
T/F: Cytokines are soluble but not membrane-associated
FALSE
T/F: Cytokines can be produced by non-immune cells such a epithelial and vascular cells in response to PAMPs
TRUE
What are the major groups of cytokines?
1. Pro-inflammatory
2. Anti-inflammatory
3. Interferons
4. Interleukins
5. Chemokines
6. Growth Factors
What are the clinical signs of Inflammation?
Heat
Pain
Redness
Swelling
What serum proteins accumulate in the tissue during inflammatory response?
Complement Proteins
Antibodies
Cytokines
Chemokines
Growth Factors
Albumin
Which innate immune cell generates dif. kinds of pro-inflammatory cytokines?
Macrophage = one of the 1st cells on the scene (like channel one news...)

Produces a lot of inflammatory cytokines
Why is fever good for us?
Virus/Microbes don't replicate well at high temperatures, but our immune cells DO work well at high temps.

Immune cells like it HOT.
What are the actions of Macrophage-derived cytokines TNF-alpha, IL-1, IL-6? (=occur during inflammation)
Liver - Acute-phase proteins, like C-reactive protein, mannose-binding lectin --> Activation of Complement Opsonization

Bone Marrow/Endothelium - Nuetrophils --> Phagocytosis

Hypothalamus - Inc. Body Temp. --> Dec. Viral and Bacterial Replication

Fat/Muscle - Protein/Energy mobilization --> Inc. Body Temp. --> Dec. Viral and bacterial replication
What proteins are part of the Acute-Phase Response?
Protein C (CRP, Complement Activation) and MLB (Initiates Lectin Complement pathway)

Goal: Get rid of infection
Bacteria induce macrophages to produce what?
IL-6, which acts on hepatocytes to induce synthesis of acute-phase proteins like mannose-binding lectin, fibrinogen, and C-reactive protein
What does C-Reactive Protein do?
Binds phosphocholine on bacterial surface acting as opsonin and a complement activator
What does mannose-binding lectin do?
Mannose-binding lectin binds carbs carbs on bacterial cell surface acting as opsonin and complement activator
When they sense microbial products, what pro-inflammatory cytokines do macrophages secrete?
IL-6
TNF-alpha
IL-1beata
CXCL8 (=IL-8)
IL-12

ALL Increase permeability of vasculature so things can get to an area of infection
What does IL-12 do?
Inc. Th1 cells
Dec. Th2 cells
What does IL-8 (=CXCL8) do?
Recruits Neutrophils and Basophils to site of infection
What does IL-1beta do?
Activates:
Vascular Endothelium
Lymphocytes
Local Tissue Destruction
Increases Access of Effector Cells

Systemic:
Fever
Production of IL-6
What does TNF-alpha do?
Activates:
Vascular Endothelium
Inc. Vascular Permeability = inc. entry of complement cells
Inc. Fluid Drainage to Lymph Nodes

Systemic:
Fever
Mobilization of Metabolites
Shock
What does IL-6 do?
Only Systemic:

Fever
Induces Acute-Phase Protein Production by Hepatocytes
Can inflammatory effects of cytokines be bad?
Yes
What is Sepsis?
Bacterial infection in blood resulting in toxic effects

Usually caused by GRAM NEGATIVE - bacteria
What is Septic Shock?
Caused by systemic release of cytokine TNF-alpha after bacterial infection of bloodstream

Dec. blood volume, Hypoproteinemia, Neutropenia, Neutrophilia

Dec. Blood Volume = Causes collapse of blood vessels and multi-organ failure

Frequently Fatal
What innate molecules in our bodies causes Septic Shock
TNF-alpha (=released by macrophages)

Can lead to death when released systemically
What are two Type 1 Interferons
IFN-alpha

IFN-beta
What does IFN-alpha do?
Induces antiviral response and increases MHC class I expression on surface of infected cells

Produced by Leukocytes and Dendritic Cells
What does IFN-beta do?
Same as IFN-alpha: Induces antiviral response and increases MHC class I expression on surface of infected cells

Produced by Fibroblasts and Virus-infected cells
What is the major Type 1 Interferon Producing cell in the body?
Plasmacytoid Dendritic Cell

=Good at detecting viruses in blood and initiating the appropriate adaptive immune response
Detection of PAMPs stimulates what?
Production of Type 1 Interferons (INF-alpha and IFN-beta - can shut down viral infection)
What are the actions of Type 1 Interferons (=INF-alpha and INF-beta)?
1. Induce resistance to viral replication in all cells (neighboring cells)
2. Inc. expression of ligands for receptors on NK cells
3. Activate NK cells to kill virus-infected cells
What are the Pro-inflammatory Cytokines? The Ant-Viral cytokines? Adaptive Immune Cell Cytokines?
Pro-inflammatiory Cytokines = IL-1, IL-6, IL-8, IL-12, TNF-alpha

Anti-Viral Cytokines = Type 1 Interferons IFN-alpha and IFN-beta

Immune Cell Cytokines = IL-2, IFN-gamma
How does an Antigen Presenting cell drive differentiation of CD4+ cells into Th1, Th2, Th17, Tregs?
Antigen Presenting Cells produce cytokines that drive differentiation of CD4+ cells into Th1, Th2, Th17 and Tregs
What does the Adaptive immune cell cytokine IL-2 do?
Activation and proliferation of T cells
What does the Adaptive immune cell cytokine IFN-gamma do?
Activates macrophages, NK cells, Neutrophils and Dendritic Cells
When an antigen presenting cell produces IL-12, what does that induce Th1 cells to do?
Makes Th1 Cells Produce IL-2 and IFN-gamma which promote T Cell Proliferation and Macrophage activation
When an Antigen producing cell produces IL-4, what does that induce Th2 cells to do?
Makes Th2 cells produce IL-4, IL-5, IL-6, IL-10, IL-13

= INC. Humoral Immunity
= Eosinophil-Mediated Destruction of Pathogens
T/F: CD40L (=CD40 Ligand) on Th1 cells helps to activate macrohages resulting in enhanced killing
TRUE
What does CD40L on Th2 cells help to do?
Activate B Cells
When an Antigen producing cell produces IL-6 and TGF-beta, what does that induce Th17 cells to do?
Recruit Neutrophils
Inc. Inflammation

NOTE: BOTH IL-6 AND TGF-beta have to be present to Induce Th17 cells
Without the presence of IL-6, what does TGF-beta alone do when produced by antigen presenting cells?
Makes Treg produce IL-10 and TGF-beta which suppress adaptive immune responses
T/F: TGF-beta in ABSENCE of IL-6 will give us a regular T cell (Treg)
TRUE
What are the two major immunosuppressive and anti-inflammatory cytokines?
TGF-beta and IL-10
What does TGF-beta do?
Suppresses antigen-specific Th1 and Th2 cells

Blocks dendritic maturation

Induces isotype switching to IgA

Role in generating Tregs and is produced by Tregs
What does IL-10 do?
Blocks differentiation of Th2 cells

Inhibits IFN-gamma and IL-2 production

Inhibits IL-4 and IL-5 production from Th2 cells

Inhibits Inflammatory cytokine production from phagocytes
List some cytokines that induce isotype switching
IFN-gamma = induces switching to IgG1

IL-4 and IL-3 Induce switching to IgE (activates mast cells ie role in inflammation)

TGF-beta induces switching to IgA (mucosal immunity)
Hematopeiesis is the generation of?
Cells of the blood including RBCs, WBCs and Platelets
T/F: RBCs, WBCs, and platelets all derive from pluripotent hematopoietic stem cells
TRUE
What is the hematopoeitic stem cell marker?
CD34
What are the hematopoetic cytokines?
G-CSF
GM-CSF
What does the hematopoeitic stem cell marker G-CSF do?
Promote proliferation of granulocyte precursors (neutrophils, eosinophils and basophils)
What does the hematopoeitic stem cell marker GM-CSF do?
Growth and differentiation of dendritic cells, monocytes, macrophages, granulocytes