• 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

1/173

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;

173 Cards in this Set

  • Front
  • Back
Where are naive lymphocytes found?
in circulation, lymph nodes, spleen

Where they have never encountered an antigen
Where are activated lymphocytes found?
At the peripheral site of infection where they have encountered an antigen
Antigen
Part of the pathogen that you immune system can recognize
Types of immune responses
Antigen specific

Antigen non-specific
What are the advantages/disadvantages of Antigen Specific immune responses?
Advantage:
Can differentiate between foreign and self (response will not attach host cell)

Disadvantage:
Response is slower (5 days)
Which immune cells recognize foreign antigens
Only B and T lymphocytes
T-Lymphocytes
Type Types: Alpha-Beta and Delta-Gamma?

AB cells
-CD4 effector cells
-CD8 cytotoxic cells
-determines what type of pathogen the T--cell will recognize (intra vs extra cellular)

DG cells
-live in tissues
-Involved in periapical lesions
-Look like T-cells in the blood but dendritic cells in the tissues
B-Lymphocytes
Two Types: Conventional cells and CD5 cells

Cells secrete antibodies that will recognize foreign antigens

Conventional cells
-majority of cells

CD5 cells
-Minority of cells
-Live in tissues
-Work faster than Conventional B-cells
What is the advantage/disadvantage of Antigen Non-Specific Immune Response
Advantage:
Response is faster

Disadvantage:
Cannot differentiate between foreign and self (may also attack host cells)
What type of cells perform Antigen Non-Specific Immune Response
Phagocytes (monocytes, macrophages, dendritic cells)

Granulocytes (PMN/neutrophils, basophils, eosinophils, mast cells)

Natural Killer Cells (relative of T-cell)
Which cells are the first to arrive at the site of bacterial infection?
Neutrophils/PMN (granulocyte)
Types of Lymph Organs
Primary Organs - where cells develop

Secondary Organ - where cells spend most of their life
Primary Organs
Where cells develop

Includes:
Bone Marrow

Thymus

Start from a common progenitor int he bone marrow

T-cells leave bone marrow and migrate into thymus to mature while other cells remain in bone marrow
Secondary Organ
Where the cells spend most of their life. Function to unite T-cells and B-cells with stuff from periphery

Includes:
Bone Marrow

Spleen

Tonsils & adenoids

Lymph Nodes & Vessels

Bronchus (BALT, MALT)

Gut (GALT, MALT)

Urogenital lymphoid tissue, Peyer's patch
Function of lymph vessels
Drains fluid from peripheral tissues into the lymph nodes

Pathogens travel with lymph vessels into nearest lymph node where you immune system can respond
How do cells enter and exit the thymus
There is NO lymphatic vessels so cells enter/exit via blood circulation
Structure of Thymus Gland
Dense cortex
Less dense medulla

No lymphatic vessels

No reticular fibers
Involution of Thymus Gland
Beings at puberty

Most of the glandular tissue is replaced with fat

T-cells no longer develop fast therefore lifespan is very long

90% of T-cells are present when you hit puberty

Death of T-cells cannot be replaced
Cells of the Thymus Gland
Thymocytes - becomes T-cells

Dendritic cells - support the thymocyte as they develop

Hassall's Corpuscles - unique only to thymus (unknown function)

Macrophages
Dendritic cells
AKA Reticular Epithelial cells

Support the thymocyte cells in the Thymus develop into T-cells
Thmocytes
Cells of the Thymus gland that eventually become T-cells
Function of Lymph Nodes
Work horses of immune system

Unite B and T-cells with pathogens coming in from the periphery
Structure of Lymph Nodes
High endothelial venules in the cortex

Dense cortex
Less dense medulla with cellular cords and empty sinuses

Has afferent and efferent lymphatic vessels
Afferent Lymphatic Vessels
Drain peripheral pathogens into the nearest lymph nodes
Efferent Lymphatic Vessels
B and T-cells exit this pathway to go out to the peripheral site of infection to defend against those pathogens
Diapedesis
Movement of cells from periphery into the lymph nodes

Facilitated by the puffy, big spaces within lymph nodes
Cells present in Lymph Nodes
B-cells - Live in cortex

T-cells - live in paracortex

Dendritic cells & Macrophages - live in follicles and paracortex

Reticular cells with fibers
B-cells in Lymph Nodes
Live in the outer cortex of Lymph Nodes

Unactivated B-cells are in primary follicle

Activated B-cells are in secondary follicle
T-cells in Lymph Nodes
Live in paracortex, the zone in between the outer cortex and the inner medulla
Structure of Spleen
Consists of red pulp and white pulp

Red pulp functions as a filter to clean out old RBC and debris

White pulp functions as a lymph node only during systemic infection

Cords have reticular cells

Sinuses have reticular fibers

Blood infections can be cleared out through the spleen since it doesn't go through lymph nodes
Red Pulp
Most of the structure of the spleen

Less dense

Stains red

Lots of RBCs and macrophages

Functions as a filter to clean out old RBC and debris from the immune response
White pulp
Area of Spleen organized in areas surrounding capillaries

More dense

Stains blue

Lots of WBCs, macrophages, and dendritic cells

Function like a lymph node only in the event of a systemic infection.
Which organs are encapsulated and have septa?
Spleen, thymus and lymph nodes

(Tonsils are variable)
Which organs have cortex and medulla?
Thymus

Lymph nodes

(Spleen and Tonsils do not)
Which organs have sinuses and cords?
Spleen

Lymph nodes

(Thymus and Tonsils do not)
Cells of the Spleen
B-cells - found in follicular and germinal regions in white pulp

T-cells - found in Periarteriolar Lymphatic Sheath (PALS) of white pulp

macrophages - in both white and red pulp

Dendritic cells - in white pulp only
Why do you need antibiotics for surgery but not for exfoliation/extraction of teeth?
Enclosed environments like your gut don't have the same full-blast immunity
Function of Tonsils
They are a reservoir for bacteria

Act like lymph nodes to maintain contact between bacteria and functional memory immune cells

The reservoir of bacteria can also contribute to re-populating the pathogens that cause periodontal disease
Types of Tonsils
Palatine

Pharyngeal

Lingual
Palatine Tonsils
Bilateral (2)

Located in oropharynx

Stratified Squamous Epi

Partial thick capsule

10-20 crypts/tonsil

Crypts hold aerobic and anaerobic bacteria
Pharyngeal Tonsil
AKA Adenoid (when inflamed)

1

Located in nasopharynx

Startified Squamous Epi - less epithelium

Partial thin capsule

No crypts, but has folds

Lots of Lymphocytes
Which tonsil does not have crypts
Pharyngeal

It has folds
Lingual tonsil
Numerous

Located at root of tongue

Stratified squamous epi

No capsule

1 crypt per tonsil
Which tonsil does not have a capsule
Lingual tonsils
Which tonsil has numerous crypts
Palatine tonsils - 10-20 per tonsil
Associated Lymphoid Tissues
Discrete lymph tissues that exist in any area that is exposed to the outside environment

Absent in the stomach (stomach acid takes care of it)

Absent in skin (outer keratin layer provides barrier)

B-ALT
G-ALT
M-ALT
BALT
Associated lymphoid tissue int he respiratory tract
GALT
Associated lymphoid tissue in the GI tract
MALT
Associated lymphoid tissue in the mucosa
M-Cell
Cell of MALT/GALT

no known function

Takes in all kinds of particles

Forms tight junctions with surrounding enterocytes (intestinal absorptive cell)

Food particles get transported through M-cell and induces tolerance to food

Polio, shingella, salmonella causes food poisoning by invading M-cells and entering through tight junctions into enterocytes

Houses B and T-cells to fight pathogens
Which organ do WBC NOT travel to?
Thymus
Which organ is never involved in the immune response
Thymys b/c the cells there are not yet mature to fight pathogens
Why doesn't the thymus gland have lymphatic vessels
Because there's no point in sending pathogens to immature T-cells that can't handle them
Homing
Cells tend to go to the place where they got activated

Homing is controlled by cell surface proteins
Which organs have lymphoid follicles
Lymph Node

Spleen

Tonsils

(Thymus does not)
Which organ does NOT have a cortex and medulla
Spleen

Tonsils
Which organ does NOT have Lymphoid follicle
Thymus
Which organ does NOT have cords and sinuses
Thymus

Tonsils
Which organ does NOT have a capsule
Tonsils (variable)
Which organs have a hilum
Lymph Node

Spleen

(Thymus and Tonsils do not)
Which organs do NOT have a hilum
Thymus

Tonsils
Which organs have B-cells
Lymph nodes - Follicles in outer cortex

Spleen - Peripheral white pulp

Tonsils - Follicles
Which organ does NOT have B-cells
Thymus
Which organ has T-cells
Thymus - whole organ

Lymph Node - Inner Cortex (paracortex)

Spleen - Periarterial Lymphatic Sheathe

Tonsils - between follicles
Which organ does NOT have T-cells areas
All organs do
What is the unique structure for Thymus
Epithelial cells/Hassall's corpuscles
What is the unique structure for Lymph Nodes
Afferent lymphatics

Follicles adjacent to capsule
What is the unique structure for Spleen
Central arteriole
What is the unique structure for Tonsils
Follicles adjacent to epithelium
Antigen Non-Specific Cells
Kill by eating or releasing killer granules

Ex: phagocytes, granulocytes, natural killer cells
IL-1
Macrophage Cytokine

Activates vascular epi

Increase access of effector cells

Activates naive T-lymphocytes

Causes local tissue destruction

Produces fever

Upregulates IL-6
IL-6
Macrophage Cytokine

Upregulates antibody production

Increases lymphocyte production

Assists in B-cell activation

Produces fever
TNF-a
Macrophage Cytokine

Activates vascular epi

Shuts down venous return to increase fluid draining to lymph nodes (causes shock)

Produces fever

Mobilizes ROS metabolites

Activates neutrophils (with IL8)

Activates dendritic cells
IL-8
Macrophage Cytokine

A chemokine

Steers other immune cells to the site of antigenic challenge

Activates neutrophils (with TNGa)
IL-12
Macrophage Cytokine

Activates NK cells

Causes differentiation to TH1 T-cells
IFa/IFb
Macrophage Cytokine

Inhibits protein synthesis

Inhibits DNA replication in virus-infected cells

Increases MHC class I expression and antigen presentation in all cells

Activates NK cells against virus
IF-gamma
TH1 Effector T-cell cytokine

Induces respiratory burst enzymes to generate activated destructuve macrophages

Increases activated CD8 T-cell activity

Increases activated NK cell activity

Causes B-cells to swtich to IgG Isotype
IL-2
TH1 Effector T-cell cytokine

T-cell growth factor leading to clonal proliferation when activated by dendritic cell costimulation

Induces apoptosis in effector T-cells
LT
TH1 Effector T-cell cytokine

Inhibits B-cells

Kills T-cells

Activates macrophages

Activated PMN

Kills tumor cells
FasL
TH1 Effector T-cell cytokine

Induces apoptosis in Fas bearing cells
Complement
Proteins found in serum that are usually inactive and when activated can go into 2 different pathways

-classical (antibody activated) and alternative (self activated)
Classical Complement Pathway
Antibody binds to specific antigen on pathogen surface

Antibody activation

Involves 9 complement serum proteins
Alternative Complement Pathway
Pathogen surface creates local environment conducive to complement protein activation

Self activation

Involves 8 complement serum proteins
What are the first cell responders to infection?
Dendritic cells- always lie in tissue

Macrophages
C3b
Keeps bacterial load in check during the next 5 days as adaptive immune response kicks in

It Opsonizes bacteria to facilitate recognition and phagocytosis by neutrophils

Transports RBC to spleen for filtration

Activated B-cells
Anaphylotoxins
Fragments of complement serum proteins (C2a, C3a, C4a, C5a)

C5a > C3a > C4a > C2a in effectiveness

Recruit neutrophils and macrophages by activating expression of LFA-1 and CXCL-8 receptors on WBCs

Alter vascular epi making them leaky allowing WBC to migrate out of blood vessels into site of infection
How to fight off viruses?
Interferon a and B (IFa, IFb) - produced by infected hold cell to disrupt viral replication

Natural killer cells
Interferon A and B
Produced by an infected host cell to disrupt viral replication
Natural killer cells
Looks for absence of MHC-1 on cells that have been removed from viruses and kills those virally-infected host cells
What complement initiates the polymerization of the Membrane Attack Complex
C5b
What complement pokes a hole in the bacterial membrane
When C8 binds to C5b-C6-C7
What complement forms a cylindrical pore in the bacterial cell membrane causing the bacteria to explode and die?
When 10-15 units of C9 binds to C5b-C6-C7-C8
Order of important/effectiveness of anaphylotoxin A-pieces
C5a > C3a > C4a > C2a
Glomerulonephritis
When the spleen cannot filter out antibody-antigen complexs on RBC

They end up getting trapped in the glomerular capillary beds
Deficiency in which complements cause disease?
Deficiencies C3b and C5a are associated with severe diseases

Anything further down in complement is not crucial to survival
C1 Inhibitor
Soluble factor

indiscriminately inactivates complement on both the bacteria and host cell to turn off complement cascade

Takes away C1 to prevent cleavage of C4 and C2
C4 binding protein
Soluble Factor

indiscriminately inactivates complement on both the bacteria and host cell to turn off complement cascade

Dissociates C2a
Factor H
Soluble Factor

indiscriminately inactivates complement on both the bacteria and host cell to turn off complement cascade

Type of C3 Convertase Inhibitor

Inactivates C3b in the Alternative Pathway
Factor I
Soluble Factor

indiscriminately inactivates complement on both the bacteria and host cell to turn off complement cascade

Type of C3 Convertase Inhibitor

Inactivates C3b in the Alternative pathways
and
Inactivates C4b in the Classical Pathway
Soluble Factors
indiscriminately inactivates complement on both the bacteria and host cell to turn off complement cascade

Includes:
C1 Inhibitor
C4 binding protein
Factor H
Factor I
Membrane Bound Factors
Specifically inactivates complement only on the host cells

Includes:
DAF
MCP
Cd=59
DAF
Membrane-bound Factor

Specifically inactivates complement only on the host cells

Dissociates Factor-Bb to inhibitor the formation of C3 Convertase in the Alternate Pathway

Makes it ok for C3 to spontaneously cleave without hurting your host cells
MCP
Membrane-bound Factor

Specifically inactivates complement only on the hold cells

Dissociates Factor-Bb to inhibitor the formation of C3 Convertase in the Alternate Pathway

Makes it ok for C3 to spontaneously cleave without hurting your host cells
Cd-59
Membrane-Bound Factor

Specifically inactivates complement only on host cells

Prevents the formation of C9 pore structures
How do WBC travel through vessels?
Rolling adhesion

Tight binding

Diapedesis - Cells get sucked through between endothelial cells out of blood vessels

Migration
Rolling Adhesion
Weak binding between endothelial Selectin molecules and WBC S-Lex receptors allows the WBC to roll along with the force of blood flow
Tight binding
CXCL-8 and ICAM-1 on the endothelium surface induces a change in the endothelium that allows stronger bonds so that the WBC is stopped and can no longer roll
Diapedesis
WBC gets sucked through between endothelial cells out of the blood vessel
Migration
WBC follows the CXCL-8 gradient being secreted at the site of infection
How do neutrophils, macrophages, dendritic cells recognize bacteria once at the site of infection?
They have receptors that can recognize things specific to bacteria like:
Endotoxin
Mannose
n-formylmethionine
FC-receptors
complement
Once macrophages recognize bacteria at the site of infection, what do they do?
They secrete cytokines that function to:

activate innate immune response

activate adaptive immune response
Dendritic cells
Immature in the tissues until activated by TNF-a
How are antigens presented to T-cells
Foreign antigens are presented on Major Histocompatibility Complex (MHC)/ Human Leukocyte Antigen (HLA) docking proteins on the surface of dendritic cells
MHC
Major Histocompatibility Complex

Same as Human Leukocyte Antigen (HLA)

They are proteins on the surface of dendritic cells that recognize foreign antigens
CD4 T-cells
Looks for extracellular (bacterial) peptides on HLA-2 docking proteins
CD8 T-cells
Looks for intracellular (viral) peptides on HLA-1 docking proteins

Lyse the host cell via apoptosis
Virus gets killed too because it lives inside the lysed host cell
HLA-1
AKA MHC-1

Human Leukocyte Antigen-1 protein made by most nucleated host cells (except RBC-no nuclei and neurons)

Composed of heavy and light chains

Activates only CD8 T-cells

Acquires peptides in ER

Heavy chains binds to CD8 co-receptors
HLA-2
AKA MHC-2

Human Leukocyte Antigen-2 protein made by antigen presenting host cells (APC) only

Ex: Dendritic cells, B-cells, macrophages

Composed of alpha and beta chains

Activates only CD4 T-cells

Acquires peptides in vesicles

Beta chain binds to CD4 co-receptors
what is binding specificity of the T-cell receptor determined by?
T-cell receptor does not determine binding to HLA-1 or HLA-2

Binding specificity is determined by CD4 and CD8 co-receptors
Which cells make MHC/HLA-1 proteins?
Most nucleated host cells

Except RBC - they dont have nuclei

Except Neurons - you don't want to be killing these cells because they can't regenerate
Which cells do not make MHC/HLA-1 proteins?
RBC because no nuclei

Neurons because you don't want to be killing these cells
Which cells make MHC/HLA-2 proteins?
Made only by antigen presenting host cells (APCs)

Ex:
Dendritic cells
B-cells
Macrophages
What is the only APC that presents antigens to activate CD4 and CD8 T-cells
Dendritic cells

Both B-cells and Macrophages present antigens only to activate itself
MHC/HLA-1 Protein Synthesis
Makes heavy and light chains by getting building blocks from pool of degraded peptides in ER

If healthy, the proteins will be self peptide

If virally infected, the proteins will be viral peptides

HLA-1 gets transported through cell in Golgi vesicle to be exocytosed on to host cell membrane surface

HLA-1 can now bind to CD8 T cell receptors on the cell membrane
MHC/HLA-2 Protein Synthesis
Dendritic cell must first bring bacteria inside through phagocytosis

Bacterium gets degraded into peptide segments

Invariant chain chaperone protein prevents HLA-2 from getting peptides from ER and instead signals it to find a phagosome to acquire bacterial peptides

If healthy, CLIP segment remains attached to HLA-2 blocking acquisition of vesicle peptides

If bacterially infected, HLA-dm removes CLIP allowing HLA-2 to acquire peptides in veiscles

HLA-2 then gets exocytosed and can now bind to CD4 receptors on dendritic cell membrane
CLIP
Segment that remains attached to HLA-2, preventing acquition of vesicle peptides to form, unless the dendritic cell is bacterially infected
MHC/HLA-dm
Removes CLIP segment for HLA-2 allowing it to assemble using peptides in vesicles

Only removes CLIP when bacterially infected
Invariant Chain
Chaperone protein in APC cells that prevents HLA-2 from acquiring peptides in the ER

Signals to HLA-2 to go find a phagosome where it may be able to acquire its bacterial peptides
How is antigen recognition maximized
Polygenism - HLA1-A, HLA1-B, HLA1-C, etc

Polymorphism - HLA1-AA, HLA1-Aa, HLA1-aa

Multiplication - peptide binding groove binding number different peptides

60,000 possible peptides can be recognized by B-cells and T-cells

This is how MHC/HLA polymorphism and polygenism ensures that T-cells can recognize any possible antigen on any kind of foreign invaders
How are MHC/HLA peptide binding grooves able to recognize and bind so many different peptides?
Anchor Residues - specific hydrophobic AA at specific locations on the peptide

MHC/HLA-1 has anchor residues at the ends

MHC/HLA-2 has anchor residues in the middle
T-cell Receptor
Specific for both allele and the peptide

Both must be a correct match in order to activate the T-cell
Which cells perform antigen processing and presentation
Dendritic cells
How do B-cells get activated?
1. B-cell antibody receptors bind to antigens on dendritic cell

2. Receptor mediated endocytosis causes B-cell to phagocytose the bacteria and produce HLA-2

3. B-cell leaves primary follicle (outer cortex) of lymph node looking for T-cell in paracortex of lymph node

4. B-cell presents its antigen peptide to a receptor on the TH2/CD4 cell

5. CD-40 ligand on activated TH2/CD4 cell binds to CD-40 receptor on unactivated B-cells

6. TH2/CD4 cell cytokines bind to B-cell receptors

7. B-cell returns back to primary follicle and undergoes clonal proliferation in the secondary follicle (outer cortex)

8. daughter B-cells under somatic hypermutation resulting in variability in antibody binding capabilities

9. Undergo further rounds of proliferation and hypermutations to produce progressively stronger affinities

10. Strongest daughter B-cells differentiate into Plasma cells to produce antibodies at the site of infection

11. At the end of infection, some daughter B-cells differentiate into Memory Cells to keep you protected for life

12. Isotype switching of IL4 to IgE, and IL-10 to IgA
Antibody
Antigen specific receptor of B-cells

They can recognize almost any stucture such as proteins, lipids, carbs, etc, on the bacterium (T-cells can only recognize proteins presented on MHC cells)
IgM
COMPLEMENT ACTIVATION

Activates complement by binding bacterium

First kind of antibody to be secreted
IgE
MAST CELL SENSITIZATION

Mast cells have high affinity receptors for IgE

Causes mast cells to release all its granules

Leads to allergic/asthma symptoms - coughing, sneezing, itching etch

Not found free in serum - always bound to mast, basophils, eosinophils
IgD
B-CELL ACTIVATION

Only function is to activate B-cell

Does not get secreted into serum
Which isotypes do Naive B-cells have
IgM and IgD
Which isotypes do Activated B-cells have
Isotype switching occurs in daughter B-cells and only happens after B-cell is activated
Clonal deletion
Form of tolerance

If B-cell makes receptor that recognizes self, the B-cell will die by apoptosis
Anergy
Form of tolerance

If B-cell receptor binds to a soluble antigen, the B-cell will become anergic (permanently deactivated)
Central Tolerance
When tolerance occurs in the thymus or bone marrow (developmental organs)

Like clonal deletion or anergy
Peripheral tolerance
When B-cells are anergized (deactivated)
Maturation
If there is no self-reaction, mature, but naive B-cells will have IgD and IgM on its cell surface and migrates into circulation to flow from lymph nodes to spleen.

If B-cell does not find antigen, it dies
CD5 B-cell
AKA B1 B-cells

Small part of total B-cell population (majority is Conventional B-cells)

Lives in tissues

Does NOT need T-cell activation

Secretes IgM antibodies which activates classical and alternative complement

Recognizes bacterial carbs
How are T-cell receptors made
Gene arrangements and nucleotide splicing occurs in the thymus cortex

It maximizes diversity in receptor recognition abilities

T-cell receptors that cannot bind to HLA receives no CD3 survival signals and causes T-cell to die via apoptosis

T-cell receptor that can bind to HLA receives CD3 survival signal
Positive Selection during T-cell Development
T-cell needs to demonstrate that it can bind well enough to HLA molecules and peptides which will give it a survival signal from CD3

T-cell will receive survival signal if it can recognize self peptides

High affinity will prevent apoptosis

Type of HLA that T-cell receptor binds determines if that T-cell becomes CD4 or CD8 cell
Negative Selection during T-cell Development
T-cell needs to demonstrate that it won't bind to HLA too well

Don't want T-cell to bind too tightly to host peptides and kill the host

Test to see if T-cell Receptor binds too tightly to self protein

High avidity (the sum of strength of binding molecules to one another) induces apoptosis

Negative selection induces apoptosis
Type I diabetes and Positive/Negative Selection
They don't do positive and negative selection very well

Therefore some T-cells will attack islets of Langerhans decreasing production of insulin
How are dendritic cells so good at activating T-cells
They have evolved so that they can be infected by all types of viruses (that means dendritic cells can put viral peptides on HLA-1)

They can secrete IFa and IFb to prevent viral replication and consequential destruction of itself

They have a shuttle mechanism to transport degraded viral peptides into vesicles (allowing Dendritic cells to put viral peptides on HLA-2)

Therefore can activate both CD8 T-cells (HLA1) and CD4 T-cells (HLA2)

Can secrete DC-CK to recruit both types of T-cells
DC-CK
Dendritic Cell Chemokine


Secreted by activated dendritic cells that have arrived at the lymph node

Recruits both types of T-cells
How do T-cells get activated?
1. dendritic cells are still immature in the tissues

2. During infection, TNFa is secreted by macrophages signalling dendritic cells to diapedesis out of blood vessels

3. Dendritic cells become activated in the paracortex of lymphnodes allowing them to secrete DC-CK and present HLA molecules

4. Naive T-cells cruise lymph nodes and are activated by antigens present on dendritic cells

5. T-cell LFA-1 binds to dendritic ICAM-1 to allow T-cell to roll along surface of dendritic cell to scope out correct HLA-peptide

6. If there is a match, T-cell stays in lymph node to get activated via phosphorylation signals (CD3 and co-stimulatory signal)

7. If both signals given successfully, T-cell secretes IL-2, and causes clonal proliferation
What are the phosphorylation signals that get T-cells activated in the lymph node?
1st signal - CD3
Changes conformation to allow tight binding

2nd signal - co-stimulation
Dendritic cell binds to T-cells CD-28.

Causes T-cells to secrete IL2 leading to clonal proliferation creating 10,000 daughter T-cells
What occurs if there is no co-stimulatory signals produced by the APC cells
T-cell gets anergized to prevent host cell destruction
TH1 cells
Type of CD4 T-cell

Leaves the lymph node to drive adaptive immune response at the site of infection
TH2 cells
Type of CD4 T-cell

Stays in lymph node to activate B-cells so that you can produce antibodies
How does the body separate naive B/T cells from Activated B/T cells fro the site of infection?
Activated cells switch their receptors from Selectin to become Integrin

Activated endothelium switch their molecules from Adressins to become Adhesin in response IL2

Naive T-cells cannot leave circulation because they have L-Selectin receptors that can only bind to S-Lex on un-activated endothelium

Activated T-cells are forced to site of infection because they have VLA-4 receptors that can only bind to VCAM on activated endothelium
Activation of macrophages
Macrophages cannot degrade bacteria to their fullest killing potential until TH1/CD4 cells leave the lymph node

TH1 cells receptor binds to Macrophage's HLA2

TH1 cell's CD40 ligand binds to macrophages CD40 receptor (coactivation)

TH1 cell secretes IF-gamma that binds to macrophages IF-gamma receptor

Macrophage can now effectively degrade and can secrete ROS to kill un-eaten bacteria
Granuloma
Formation of a wall of T-cells to try to isolate the un-eaten bacteria.

This occurs when there is no activation of macrophages

Ex; Tuberculosis
Tuberculosis
Wall of lymph tissue forms around the alveoli and interferes with breathing

macrophages cannot effectively lyse TB bacterium do to resistant coating and the T-cell has to surround the un-eaten bacteria
Foamy Macrophages
Activated macrophages that have eventually exhausted their degrading capabilities and come to the end of their life

Ends is apoptosis
Apoptosis of Macrophages
Occurs when they become Foamy

TH1 FAS-ligand binds to macrophages FAS-receptor to induce apoptosis

Cell membrane dissolves last

Everything inside the macrophage degrades before cell membrane explodes
IF-gamma
Main TH1 cytokine

Coordinates all the activities involved in fighting an extracellular infection

Macrophages are coactivated w/ CD40-ligan

NK cells are made more efficient

CD8 T-cells are made more efficient

B-cells switch isotypes to IgG giving NK and macrophages their targets

Epithelial cells upregulate HLA-1 production and HL2 expession
NK cells
are not antigen-specific because they don't have antigen receptors. They can only recognize antigens when bound to IgG antibodies

Only need IL-12 to get activated (IL-12 secreted by macrophage)

Also secrete IF-gamma keeping TH1 and macrophage activity up and makes CD8 cells more effective in fighting intracellular infections
How do effector CD8 T-cells kill cells that have been intra-cellularly infected?
Two ways:
Perforin and Granzyme causes infected cell to go through Capase Pathway leading to apoptosis
and
FAS-Ligand (apoptosis)
Perforin
Secreted by CD8 T-cells

Forms a pore structure in the infected cell
Granzymes
Secreted by CD8 T-cells

Enters the infected cell
FAS-ligand
Macrophage presents its peptide to HLA-1 to the activated CD8 T-cell

T-cells FAS-ligand binds to macrophages FAS-receptor leading to apoptosis
What drives shutting off an immune response?
The decrease in the amount of Antigens present in the body
IgG
NEUTRALIZATION, OPSONIZATION & TARGETED KILLING

Neutralization:
Actively transported into extracellular spaces

Competes against pathogens for their binding site

Cannot stop bacterial replication

Opsonization:
Bacteria gets coated to allow for easier phagocytosis by macrophages/neutrophils

Targeted Killing:
IgG binds to bacterium allowing NK cells to recognize bacteria
IgA
NEUTRALIZATION

Can be pumped across epithelial cells into lumen spaces

Primary antibody in saliva, sweat, tears, gut


Competes against pathogens for their binding site

Cannot stop bacterial replication
Fc receptors
Located on the cells that can bind to IgG

IgG - can only recognize the antibody after the antibody has bound to an antigen

IgE - Can recognize the antibody even when the antibody is unbound to an antigen
B-cell tests
If B-cell can bind to cell surface self-antigens --> apoptosis

If B-cell can bind to soluble free self-antigens --> anergic

If B-cell cannot bind to any self-antigen --> B-cell migrates out of the bone marrow and enters peripheral circulation