• 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

1/125

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;

125 Cards in this Set

  • Front
  • Back
important invasive factor
IgA protease
agglutinins and typical Ig
clump bacteria, enhance their removal- usually IgM
opsonins and typical Ig
enhance phagocytosis,
Ab coated microbes attach to Fc receptor on phagocytic cells (M/M, neuts) --- IgG most often

also- complement helps
Abs that activate complement
IgM and IgG
neutralizing Abs do what and typical Ig
AB THAT NEUTRALIZE VIRUSES OR BIOLOGIC ACTIVITY OF BACTERIAL TOXINS

IgG, IgA
lysins and typical ig
CAUSE DISRUPTION OF CELL MEMBRANES ESPECIALLY IN CONJUCTION WITH THE C SYSTEM (IgG, IgM)
anti-adherence Ig and example
PREVENT ATTACHMENT OF MICROBES TO CELLS OR HARD SURFACES. FOR EX. SIgA IN SALIVA PREVENTS BACTERIA FROM ADHERING TO SURFACES OF TEETH
what is ADCC?
antibody-dependent cell-mediated cytotoxicity

- NK binds to Fc portion of antibody bound to pathogen and NK will release contents onto infect cells
- primary way NK cells kill
active defense mechanisms at mucosal surfaces
mammary gland, gastrointestinal, respiratory, urogenital

- skin isn't active
components of GI immunology (physical// chemical/ biological barrier)
mucous- binds lectins on pathogen surface (contains defensins that poke holes)
cilliary actions
pH
enzymes/ antimicrobial peptides (lysozyme)
leukocytes at the ready incase physical barrier is bypassed
components of GI immunology (secondary)
GALT
- epithelial
- lamina propria
- mesenteric lymph nodes
oral lymphoid tissues
- waldeyer's ring
- adenoids
- palatine tonisils
- lingual tonsits
- produce secretory IgA
components of intestinal mucosal tissues
- effector cells populate healthy intestinal epithelium and lamina propria
- peyer's patches (effector cells not yet active)
- lymphoid follicles
- draining lymphatics from villi to mesenteric lymph nodes
- LP has mature, active B and T cells (75% in gut- main area
M cells
reside:
take up antigen by:
Ag transported by:
antigen then bound by:
- in columnar epithelial along with enterocytes
- endocytosis/phagocytosis
- taken in vesicles and released at basal surface
- bound by dendritic cells, present to T cells
- lead into peyer's patches (act like regular LN)
cell that can extend into gut lumen to capture antigen
- rest of its body sits in:
activated lamina propria dendritic cells

sits in peyer's patch so it can be ready to present
active effector cells in gut is constantly making _____ to protect system
IgA
effector cytotoxic lymphocytes that can integrate into the intestinal epithelia
- whatever it binds, it kills

can be alpha:beta or gamma:delta
intestinal epithelial lymphocytes
special things about mucosal immune system

- effector cells migrate:
- macrophages don't contain ______ and don't generate ___________
- function to _______ not _____ microorganisms
effector cells always migrate back to mucosa
macs don't contain TLRs --> no inflammation
restrain, not remove (we like commensal bacteria)
naive B and T cells that migrate through teh GALT (are/are not) specific for GALT
not specific until activated to become GALT specific
gut-homing effector T cells bind to ___________ to enter lamina propria

in lamina propria, they bind to _______ expressed by __________ making it __________
- bind to intestinal vascular endothelium

- bind to chemokines (CCL225) expressed by intestinal epithelium

- embed in epithelia
factor that allows gut-homing into lamina propria

factor for embedding into epithelia
L-selectin & alpha4: beta7 to MAdCAM-1 on endothelium

E-cadherin on epithelium
Abs passed to baby through breast milk
IgA- activated in gut, is specific for all mucosa, can circulate to mammary glands then give baby Gut-specific immunity
roles for secreted IgA
- can export toxins from LP when secreted
- bind and neutralize Ags internalized in endosomes
- stick in mucous, can bind to toxins, bacteria, neutralizing pathogens until it gets clipped/passes through
- can bind pathogen on M cell and take it to lymphoid tissue
- picks up Ag in endosome of M cell then take to LN tissue
why do we have two forms of IgA?
IgA1 has longer hinge region- often cleaved by proteases
IgA2 evolved with shorter hinge- present in areas with higher amounts of cleaving pathogens

IgA1 more in oral/GI/respiratory mucosa, tonsils, spleen, saliva
NOD receptors act like ________ to sense _______ in the intestinal epithelium

activated in response to ________, leads to ___________
act like TLRs to sense toxins

NFkB, leads to inflammation (cytokines + defensins)
why are gamma:delta cells places at the gut epithelial tissue?

forces ____ expression on infected cell
leads to:
- MHC independent, don't want to wait for MHC recognition to start fighting
- forces MIC expression
- apoptosis
- gamma delta cell helps repair wound
what happens when a cell loses its MHC I receptor?
means its been infected

express MIC (NK receptor)

induce NK killing
mucosal immunity against worms needs this kind of helper T cell
TH2

- don't want to activate complement or macrophages like in TH1
- want B cell activation --> IgE production
once immunized, over time IgM concentration and affinity:

over time IgG concentration and affinity
stay low

increases, better affinity through somatic hypermutation
will secondary responses produce IgM?
- no
how are naiive B cells inhibited from responding in a secondary response?

why is this important?
- B cells express FcyRIIB1
- specific IgG binds to these receptors on naiive B cells and inhibit them

1. provide feedback inhibition of an ongoing immune response
2. provide competitive mechanism to drive affinity maturation
RhoGAM prevents against this disease

disease caused by:
newborn hemolytic anemia

1) Rh- mom reacts to Rh+ from baby, makes IgM (too big to cross barrier to baby), normal baby
2) secondary Rh+ pregnancy, would have high-affinity IgG response to baby, destroy fetal RBCS
how does RhoGAM work
1) given during first Rh+ pregnancy
2) is anti- Rh+ IgG
3) since all of moms cells are naiive, it will bind to all of them and inhibit them leading to no response at all
disadvantage to inhibition of naiive B cells in specific response
pathogens evolve over time, slow changing of Ags on surface
less and less memory as Ags are mutated and specific Abs can't recognize them, all the while inhibiting naiive cells
eventually no Ags will be recognized by memory cells and you have to start all over at a primary response
how do we recognize memory T cells?
surface markers differ due to mRNA splicing

CD45RA on naiive
CD45RO on memory/effector cells
different fates of naiive T cells
straight to memory
- central memory (stay in lymphoid tissue)
- effector memory (migrate to tissues)

straight to effector cell
- then become quiescent memory cell
- most effectors die after a few days
3 types of failures of the body's defenses
1. evasion/ subversion of our defense system by pathogens
2. inherited deficiencies
3. acquired deficiencies
ways pathogens evade the immune response
1. antigenic variation (different serotypes of the same bacteria)
2. antigenic shift (dramatic)
3. latency (herpes)
difference between antigenic drift and shift
drift: generation of new viral strains- relatively mild disease epidemics

shift: something we've never seen before. no immunity to a new novel hemagglutinin
primary immunodeficiency disease characterized by lack of Abs

what symptom does it cause (especially by what kind of bacteria?)

caused by loss of this in B cells

treatment?
bruton's agammaglobulonemia

recurrent upper respiratory infections- PYOGENIC, polio

loss of btk (bruton's tyrosine kinase) --> no B cell development without it

Ig given by IV
most common immunodeficiency

what takes its place in the saliva?
selective IgA deficiency

IgM
What is DiGeorge's syndrome?

developmental or genetic?

how are B cells affected?
lack of thymus, T cell defect

developmental (due to gene deletion- not hereditary)

lower levels because you can't make TH2 T Cells
- can only have T-independent responses of IgM
characteristics of severe combined immunodeficiency disease (SCID)

how do you treat?
no T or B cells

life-threatening- bone marrow transplant only treatment
3 gene defects related to SCID
1) IL-2 Receptor gamma chain defect (component of cytokine receptors)
2) RAG 1 or 2
3) adenosine deaminase (leads to build up of metabolic products that kill T and B cells)
defect in chronic granulamoatous disease

what cannot be formed
phagocytic cells can take up microbe but are unable to break them down

- can't form O2 radicals and hydrogen peroxide
2 types of leukocyte adhesion deficiencies

both inhibit:
cause widespread:
severe ______ disease:
LAD type I: lack of LFA-1 (integrin) on neutrophils (CD18?)

LAD type II: lack of selectin LIGAND on neutrophils

inhibit diapedesis
widespread pyogenic bacterial infections
periodontal disease
loss of classical complement components leads to:

loss of MBL or alternative complement components leads to:

loss of MAC complement components leads to:
immune complex diseases (often autoimmune such as lupus)

pyogenic bacterial infections or neisseria

neisseria infections (gonorrhea, meningitis)
causes hereditary angioedema (HANE)

what does missing component normally do?

can be increased by:
symptom of concern:
lack of (or) non-functional C1INH (C1 esterase inhibitor)

normally prevents non-specific activation of C1

stress
pharyngeal angioedema- life threatening
bone marrow transplants have been successful in treating:
common gamma chain deficiency (SCID), RAG deficiency, ADA deficiency, DiGeorge’s
3 ways secondary autoimmune diseases can be caused
malnutrition (developing countries)
malignancy (problems with infections and immunosuppression caused by treatment or disease)
infections (microbe causes- like AIDs)
AIDS is caused by:

infects these types of cells:

leads to:
HIV

CD4+ TH1 and TH2 cells

opportunistic infections
when immunosuppressive therapy is used:

therapies used:

usually hinder:
those with autoimmune diseases

those receiving transplants

corticosterioids, cyclosporin A, FK506, rapamycin

stop T cell-mediated responses more than B cell responses
how does cyclosporin A work?
interferes with Il-2, prevents T-cell activation
caused by microbes usually held in check by the immune system but can be life threatening if you're immunocompromised
opportunistic infections (can be parasites, virus, bacterial, fungal)
4 types of hypersensitivites- which ones are related to antibody?
I- allergic/anaphylactic reaction
II- cytotoxic
III- immune complex disease
IV- cell-mediated delayed hypersensitivity

1st 3 are Ab related
Type I hypersensitivity Ab:
Ab increase can be induced by this factor:
antigen often called:
common allergens:
IgE
IL-4
allergen
inhaled, injected, ingested
3 features of inhaled allergens that promote priming of TH2 --> IgE release
molecular type- proteins, because they only induce T cell response
low dose- favors IL-4 producing T-cells
high solubility- allergen is readily eluted from particle
IgE half life

what does it attach to, how, and how long?

leads to:
2 1/2 days

binds to basophils and mast cells through Fc receptor with high affinity- can last for months

sensitized to allergen held by IgE, activation of mast cells/ basophils
3 effects of mast cell activation and granule release

what else can interact with mast cells and cause release of mediators?
GI tract- diarrhea, vomiting
airways- phlegm, coughing
blood vessels- edema, inflammation, increased lymph flow

anaphylatoxins
Mast Cell preformed mediators and their effects:

newly synthesized mediators and their effects:
- histamine- capillary dilation, vascular permeability, smooth muscle contraction
- ECF-A- eosinophil chemotactic factor of anaphylaxis

Newly synthesized:
Leukotrienes- similar to histamine, but even stronger
atopic/ local anaphylaxis

systemic anyaphylaxis

genetic component?
allergic rhinitis (genetic)
asthma (genetic)

anaphylactic shock= no genetic component
in chronic asthma, what cell is involved
eosinophils- damage airway
clinical features of anaphylactic shock

occurs how quickly:
symptoms in these body areas:
death can be caused by:
occur due to systemic release of:
matter of minutes
cardiovascular, respiratory, GI
death: laryngeal edema, respiratory failure, shock
histamine and leukotrienes
allergy treatment

local:
systemic:
antihistamine

epinephrine (opposite effect of histamine, in epi-pens)
IgE immediate reaction time seen/ late phase:
type III arthus reaction time:
type IV delayed reaction time:
granuloma reaction time:
<30 min/ 5 hours
5-6 hrs
24-72 hrs
21-28 days
how does hyposensitization work?
- slowly administer allergen to patient
- decrease IgE, increase IgG response
- IgG out competes IgE
3 examples of type II sensitivities
1. blood transfusion reaction: type A blood given to type B person
2. drug reactions: drug can be hapten, interact with surface protein of cells, induce Ab response
3. autoimmune hemolytic anemia: patient develops Ab against own RBCs
Abs involved in type II sensitivities

how do they work?
IgM and IgG (complement activators)

Abs bind to hapten on RBC, lead to cell lysis or phagocytosis via complement
how do type III sensitivities work?

Abs involved
Soluble Ag-Ab complexes circulate throughout the blood stream, settle in certain target organs (kidney) initiating inflammatory response

IgM or IgG
the arthus reaction is _______ induced

Abs are _________, creates Ag-AB complex

can activate ____ cells

site of rxn

will see reaction start in ________ time frame
locally

Abs are introduced (Ag already there)

mast cells --> degranulation --> inflammation

on skin

6 hrs
common causes of arthus reactions
vaccines
insect stings
oral bacteria
major organ targets for systemic type III sensitivities
kidneys, joints, heart
serum sickness

permanent?
given large does of Antigen (caused by drugs)
complexes settle in your joints(arthritis), kidney (nephritis), heart (vasculitis)
accompanied by fever

no
3 types of diseases associated with type III hypersensitivities
infectious/post infectious disease

autoimmune disease (lupus)

drug reactions (serum sickness)
how are complexes removed?
what cells destroy them?
works for which Ig:
what Ig type does this not work for?
activate complement
stick to complement receptors on RBCs
taken to KUPFFER in liver to remove/destroy

works well for IgM/IgG bc they activate complement
not good for IgA (tend to settle in organs)
type IV hypersensitivities

site:
cells often involved:
time of rxn:
sometimes involve ______ which can activate CD8 T cells
skin
TH1 --> cytokines they produce
24-78 hrs
can involve haptens soluble enough to bind MHC class I and elicit CD8 T cells response
3 examples of type IV hypersensitivities

how is it tested for?
1) tuberculin rxn
2) contact dermatitis
3) contact stomatitis (ex. cinnamon)

patch test- contains haptens that could create response, leave on for 72 hrs, see response
released by TH1 TCell in type IV
chemokines
IFN-gamma
TNF-alpha & LT (cytotoxins)
2 phases of type IV hypersensitivity
1) sensitization
- hapten makes complex with carrier
- picked up/ presented by dendritic cell

2) elicitation phase
- effector cells CD4 T cells release cytokines
- give response after 24-72 hrs
granuloma is made up of macrophages that have turned into _________ and _______

surrounded by ________

eventually causes death of cells in center of granuloma by
epithelioid

giant multi-nucleated cells

T-cells

circulation being cut-off
autoimmunity vs. autoimmune disease
response to self-antigen =not harmful

vs. loss of tolerance = harmful
5 mechanisms that contribute to immunological self-tolerance
1) negative selection in BM and thymus
2) expression of tissue specific proteins in thymus
3) sequestered Ags (no lymphocyte access)
4) suppression of autoimmune response by Treg cells
5) induction of anergy in autoreactive B/T cells
T regulatory cell surface markers

how does it work?
CD4
FoxP3
CD25

binds to same APC that autoreactive T-cell is attached to
what happens to autoreactive B cells in periphery
bind to autoantigen --> are retained in T-cell area of secondary lymphoid structures --> no T cell help --> die

peripheral tolerance
gender that autoimmune diseases are more prevalent in
females
possible causes of autoimmune diseases
1) molecular mimicry
2) breakdown in lymphatic regulation
3) microbes *association but causal not proven*
4) sequestered antigens
5) genetic
6) environmental
when antigens are shared by humans and microbes aka cross-reacting antigens

example
molecular mimicry

- body response to streptococcus
- some Abs will cross-react with myocardial antigen
- rheumatic fever
Antigens not exposed to the immune system during immunological development. How might these induce an immune response?

example?
sequestered antigens

if there is tissue damage, Ag might be exposed

sympathetic ophthalmia: damage to one eye leads to release of ntraocular protein antigens --> T cells activated --> attacks both eyes
example of a genetic autoimmune disease?

what immune cell component does it involve?
ankylosing spondylitis

HLA-B27
rheumatoid arthritis increases with age due to:
involution of thymus and less regulatory T cells
5 organ specific type II hypersensitivity diseases and causes
Hasmimoto's thyroiditis

pemphigus vulgaris: Abs to intercellular substance of the epithelium (ECM)

benign mucous membrane pemphigoid: Abs to basement membrane

autoimmune hemolytic anemia: Abs to RBCs

myasthenia gravis: Abs to acetycholine receptors
3 pathways to destruction in autoimmune hemolytic anemia
FcR positive cells in spleen

complement activation in spleen (cells with complement receptor)

complement activation and intravascular hemolysis
how can a fetus be affected by myasthenia gravis?

would a fetus be affected by Cell-mediated autoimmundisease from mom?
baby would get maternal IgG against AChR, eventually get better because the IgG wouldn't last

No- lymphocytes (Tcells) don't cross the placenta
non-organ specific (type III) disease we need to know

organs involved
systemic lupus

kidney, heart, CN system

complexes causing inflammatory response
systemic lupus erythramatosus

makes Abs against (5):

consequences (3):
DNA, ribosomes, histones, snRNP, scRNP

glomerulonephritis, vasculitis, nephritis
T-cell mediated autoimmune disease we need to know (type IV)

autoantigen:
consequence:
what type of T-cell?
type I diabetes

pancreatic B-cell antigen

Beta-cell destruction = no insulin

CD8 cytotoxic
3 rheumatic diseases caused by autoimmunity
systemic lupus erythematomus

rheumatoid arthritis

ankylosing spondylitis
definition of passive acquired immunity and two types

pros?
cons?
administration of preformed antibodies

natural: placental transfer

artificial: anti-toxins, human gammaglobulin

pros: cheap, quick acting
cons: IMMUNITY IS LIMITED
definition of active acquired immunity and two types
when exposed to the microbe or its products

natural: develops after recovering form an infection

artificial: toxoids, inactivated or killed vaccines, live attenuated vaccines
vaccine type for each disease

BCG (tuberculosis)
diphtheria
Hep B
influenza
measles
TB: live, attenuated
diphtheria: toxoid
Hep B: recombinant viral protein
influenza: inactivated virus
measles: live, attenuated
what do adjuvants do?

2 ways they do it:

ex.
non-specifically enhance the immune response

1. prolong exposure to Ag
2. initiating an inflammatory response at the site of innoculation

ex. ALUM
killed vs. live vaccines

Ag mass:
duration of immunity:
immunodeficient patient
small/ large due to proliferation of microbe
short-lived booster/ often life-long
usually OK/ risk of disseminated disease often
why is Alum important?

composed of:

mechanism of action:
it's an adjuvant used in the hepatitis B vaccine

aluminum hydroxide gel

delays release of Ag, enhanced macrophage uptake
autograft:
isograft:
allograft:
xenograft:
same person
identical twin
same species, different genetics
different species
two types of graft rejections
acute: usually mediated by T cells- takes, then turns black later

hyperacute: Ab-mediated, recipient has Abs to donor organ [immediate rejection] looks white- no vascularization at all
occurs when donor lymphocytes in the donor graft attack recipient cells

major problem for:
graft vs. host disease (GVHD)

bone marrow transplants

opposite of transplant rejection
blood transfusion reactions are examples of ______ hypersensitivity

occur when you have a _____ or ____ incompatability
type II- cytotoxic

ABO or Rh
85% of people are Rh (+ or -)

type of antibody associated with Rh

type of antibody associated with ABO typing
Rh+

IgG (has potential to cross placenta)

IgM
one of the most prevalent mucosal diseases in the world

causes an increase in what factor?
recurrent aphthous stomatitis (canker sores)

TNF-alpha
in pemphigus vulgaris, antibodies are produced against this protein
desmoglein 3
autoimmune disorder directed against exocrine glands particular salivary (________) and lacrimal glands (____________)
Sjogrens syndrome

xerostomia

keratoconjunctivitis sicca
generally caused by allogenic bone marrow transplantation

how many per year?
graft vs host disease

>4,000/year
used to detect autoantibodies in tissue (direct/indirect) or seurm (direct/indirect)
immunofluorescence for autoimmune disease
tissue = direct
serum = indirect
plasma cell dycrasias

2 examples
group of disorders where there is an abnormal proliferation of a clone Ig secreting cells (usually plasma cells) resulting in the production of monoclonal antibody (M protein)

multiple myeloma and wadenstroms macrogloulinemia
what is an M protein?

how do we test for its presence?
a monoclonal antibody (created in plasma cell dycrasias)

serum protein electrophoresis [SPE]
symptoms of multiple myeloma
bone pain, recurrent infections, mandible involved first, punched out lesions
test to differentiate between a benign granuloma from a myeloma lesion
test for kappa and lambda light chain via immunohistochemical staining

all kappa or all lambda: myeloma lesion
a mix of kappa and lambda: benign granuloma
Ig associated with wadenstroms macroglobulinemia
leads to ________ syndrome

symptoms seen:

treated with:
IgM
leads to hyperviscosity syndrome [build up of IgM in blood stream]

oozing of blood from mucosa, nose bleeding, prolonged bleeding from surgeries, disturbance of vision

treated with plasmaphoresis [thins out blood]
technique to differentiate between pemphigus and pemphigoid (type ___ sensitivities)

what are the patterns of each?
immunofluorescence: use f-Ab against auto-antibody to tell where it's localized (type II)

pemphigus: chicken wire in epithelium
pemphigoid: linear appearance at basement membrane
immunofluorescence that shows a bumpy pattern along the basement membrane is characteristic of what disease? what type of hypersensitivity?
systemic lupus erythamatosus
type III
concentration of IgM and IgD in

immature B cells leaving the BM

immature B cells after alternative splicing

mature naiive B cells

Ag activated lymphocyte
IgM only

IgM (high) IgD (low)

IgM (low) IgD (high)

IgM only
two reversible steps of B-cell receptor/Ab production
transcription activation with coexpression of surface IgD and IgM

synthesis changes from membrane Ig to secreted
when does B Cell receptor editing occur and to what?
in BM if it's autoreactive- edits the light chain
present on the surface of central memory cell that isn't on the surface of effector memory cells
CCR7
distinctive features of the mucosal immune system

anatomical features (3)

effector mechanisms (2)

immunoregulatory environment (2)
1. intimate interaction between epithelium and lymphoid tissue
2. discrete compartments of diffuse lymphoid tissue and more organized structures such as peyer's patches
3. specialized Ag uptake provided by M cells

1. activated effector T cells predominate even in the absence of infection
2. plasma cells in tissues where Abs are needed

1. active downregulation of inflammatory immune response to food/innocuous antigens
2. inhibitory macrophages and tolerance-inducing dendritic cells