Download Cell Quiz Review

Document related concepts

Immune system wikipedia , lookup

T cell wikipedia , lookup

Monoclonal antibody wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Lymphopoiesis wikipedia , lookup

Phagocyte wikipedia , lookup

Complement system wikipedia , lookup

Molecular mimicry wikipedia , lookup

Adaptive immune system wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Immunomics wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Innate immune system wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Transcript
Immuno PPT Flashcards Unit 1
Too many leukocytes is called
Too few leukocytes is called
About how many percent of all leukocytes are
basophils?
What color dye does the cytoplasm of basofils
absorb?
These granules of the basophils secrete
What is the role of histamines?
What interfere with the function of basophils?
What is a Mast Cell?
About how many percent of all leukocytes are
eosinohils?
What color dye does the cytoplasm of eosinofils
absorb?
Eosinophils cells play roles in:
The most numerous WBCs are the
What color dye does the cytoplasm of neutrofils
absorb?
Which WBCs respond first to infection?
What is the main role of the netrophils?
The nucleus of the neutrophils has how many
lobes?
How do neutrophils contribute to immunity?
How is a process of engulfing bacteria and
foreign bodies called?
What do the lysosomes do to the invaders?
When a bacterium has a capsule, it makes it
hard to phagocytize, so the neutrophil requires
what?
What do some bacteria have evolved as a
defense against phagocytosis?
Can the neutrophil or macrophage engulf
bacterias with a capsule around them?
How can neutrophil phagocytize encapsulated
bacteria?
The process of facilitation of phagocytosis is
called
When an invading bacteria has the antibody
leukophilia
leukopenia
0.5%
The basic pH dye, which is blue, so they are
called basophils (“basic dye lovers”).
histamines
Causing vasodilation; more WBCs can get to
the infection site
Antihistamines
A type of basophil that is in the tissues.
1-4%
Granules in cytoplasm absorb the pink dye
called eosin, so they are called eosinophils
(“eosin dye lovers”).
Ending allergic reactions, parasitic infections
Neutrophils
Granules in cytoplasm do not absorb either the
basic or the eosin dye, so they are called
neutrophils.
Neutrophils
Phagocytize and destroy bacteria;
Also destroy bacterial toxins in body fluids
2-6
mainly by engulfing bacteria and foreign bodies
(thorns, dirt, etc)
phagocytosis
They release the contents of their lysosomes
onto the invader, dissolving it.
opsonization by antibodies
a slippery capsule around them
No
When an antibody attaches to this type of
bacteria, the neutrophil can now grab onto the
antibody like a handle, enabling it to
phagocytize the bacteria
opsonization
antigen-antibody complex
1
Immuno PPT Flashcards Unit 1
attached to its cell membrane, the entire
structure is called
If a bacterium does not have a capsule, how can
the neutrophil destroy it?
How can the antibody destroy the bacterium by
itself?
The neutrophil and antibody work best together
when
About how many percent of all leukocytes are
monocytes?
What are the monocytes do?
What is the difference between monocytes and
neutrophils?
Monocytes use what for opsonization?
When monocytes leave the bloodstream and
enter the tissues, they are called
What characterize the dendritic cells?
What is the purpose of saving a piece of the
bacterium to present to a T-Cell?
Why are the basophils, eosinophils, neutrophils,
and dendritic cells classified as granulocytes?
Why are the monocytes not granulocytes?
Except for dendritic cells, they are all
descendants from the same precursor cell called
All of these WBCs are called
The lymphocytes and dendritic cells are
descended from a different precursor, so they
are called
How are the lymphoid cells classified?
Which WBCs are the most numerous in the
bloodstream?
Which WBCs are the most numerous in the
tissues?
How long do neutrophils live?
How long do Monocytes/Macrophages live?
How long do lymphocytes live?
What’s the Difference between Neutrophils and
Monocytes/Macrophages?
without opsonization
by popping the cell membrane
a capsule is present
about 5%
Like neutrophils, they phagocytize (eat)
bacteria, old cells, and foreign bodies.
Monocytes have more types of lysosome
enzymes than neutrophils so they are better at
killing difficult pathogens.
antibodies
Macrophages
Dendritic cells save a piece of the bacterium to
present to a T-Cell.
To activate a humoral response.
They have granules in their cytoplasm.
The monocytes do not have granules in their
cytoplasm
a myeloid cell
myeloid cells
lymphoid cells
T-Cells (three types; Helpers, Killers, and
Suppressers)
B-cells (three age groups; virgins, plasma cells,
memory cells)
Dendritic Cells (these are also granulocytes)
There are 10x more neutrophils in the
bloodstream than monocytes/Macrophages
There are more macrophages in the tissues of
the body. They are everywhere!
only a few days
few months
Lymphocytes live for years
Monocytes/Macrophages are larger and slower
than neutrophils, but they can phagocytize
2
Immuno PPT Flashcards Unit 1
What’s the Difference between Neutrophils and
Monocytes/Macrophages and Dendritic cells?
Neutrophils , macrophages, and dendritic cells
mainly function by
Lymphocytes are mostly needed to kill off
what?
All WBC’s secrete chemicals to do what?
Unlike neutrophils, macrophages have what?
What are the surface receptors?
How do the macrophages kill the bacteria?
What are the dendritic cells do?
What are the dendritic cells do with the pieces
of the bacteria’s cell membrane?
Dendritic cells present these pieces to
T cell will present these pieces to
When the B lymphocyte feels the shape of the
bacteria pieces is called
About how many percent of all leukocytes are
lymphocytes?
The most important cells of the immune system
are
How many types of lymphocytes are there?
What is the function of the two types of
lymphocytes?
Both types of lymphocytes can only act against
What are two main classes of lymphocyte?
B cells originate in the
When it has not been presented to, it is called a
After presentation, it will mature into
What is a mature plasma cell do?
After the war is over, the plasma cell will
mature into a
T cells originate in the
What are the T-cells do?
larger organisms and more of them
Neutrophils and macrophages just phagocytize
bacteria until they die. Dendritic cells
phagocytize and then take pieces of the dead
bacteria and present them to lymphocytes so a
larger immune response can occur
phagocytizing bacteria (not viruses)
body cells infected by viruses
To recruit more white blood cells to the site.
surface receptors
"recognize" the surface of the pathogen’s cell
membrane
Macrophages phagocytize the bacteria, pop
their lysosomes onto it, and dissolve it
Dendritic cells also do phagocytize the bacteria,
pop their lysosomes onto it, and dissolve it,
except they save some pieces of the bacteria’s
cell membrane
They places these pieces of bacteria on its own
cell membrane, and finds a lymphocyte to
present it to
T cell lymphocytes
B cell lymphocytes
“antigen presentation”
20–45%
Lymphocytes
2
one makes antibodies and the other engages in
direct combat with viruses and can also kill our
body cells infected with viruses
a specific foreign molecule (antigen)
B cells and T cells
bone marrow
virgin B cell
plasma cells
fights infection by producing antibodies
“queen” , called a Memory Cell
thymus gland
They attack foreign cells directly (including
3
Immuno PPT Flashcards Unit 1
B cells – start as
Plasma cells secrete
What kills the attacking cell?
In which three ways antibodies attack?
organ transplants!). They can also kill viruses
by killing the infected host cell.
virgin cells
antibodies
the plasma cell’s antibodies
-They attach to bacteria and pop the cell
membrane
-They attach to encapsulated bacteria to help
neutrophils and macrophages to phagocytize
them.
-They agglutinate (clump all over the bacteria,
binding their receptor sites so they cannot cause
harm)
Mononucleosis
When Epstein Barr virus attacks B
lymphocytes, the disease is called
How is mononucleosis characterized?
inflammation of lymph vessels (lymphangitis)
In mononucleosis, how do infected lymphocytes Infected lymphocytes have a characteristic
appear under the microscope?
scalloped
edge where they touch RBC’s
T cells – coordinate the immune response by
recruiting other white blood cells
How can T-cells destroy bacteria?
They can directly destroy bacteria by popping
their cell membrane
T cells can also directly destroy foreign cells by organ transplants
popping the cell membrane, so who are at risk?
T-cells can also kill
a host body cell that has become infected with
viruses
Do the T-cells need to phagocytize the invading No, they do not
cell?
After the T-cells kill the cell, what will happen
the macrophages dissolve the debris
with the debris?
Do the T-cells need the assistance of
No, they do not
antibodies?
Which cells attack organ transplants?
T cells
Immunosuppression drugs are designed to
inhibit the action of T cells
T cells are attacked by the
HIV (AIDS) virus
The thymus gland secrets
certain hormones
The hormones secreted by the thymus gland
T cells to become immunocompetent (makes
cause
the cells mature and start to work)
What are the main types of T cells?
Natural Killer cells, Helper T cells, Suppressor
T cells
What is the role of the Natural Killer cells?
Go out and directly kill bacteria or infected host
cells
What is the role of the Helper T cells?
Release chemicals called “cytokines” to call in
4
Immuno PPT Flashcards Unit 1
What is the role of the Suppressor T cells?
They can react to the same pathogen faster, the
next time it invades because
The study of the reactions of a host when
foreign substances are introduced into the body
is defined as
A foreign substance that induces such an
immune response in a host is
The condition in a host of being resistant to
infection is defined as
Edward Jenner found what?
The procedure of injecting powdered scabs
from someone with cowpox lesions was known
as
What is the Latin terminology for “vacca” ?
The phenomenon in which closure to one agent
produces protection against another agent is
known as
Who was Louis Pasture?
What is attenuation?
An attenuated substance is
Louis Pasteur applied this same principle of
attenuation to the prevention of
Metchnikoff, a Russian scientist, observed that
The process where white blood cells attempt to
destroy foreign objects by eating them is
Cellular immunity involves
more white blood cells of all types to join in the
war. They also present the macrophage’s
antigen to a B cell, which causes it to produce
antibodies against that particular bacteria.
Stop the immune process when it is over, and
also "tell" some plasma cells to "remember"
how to destroy that specific pathogen.
Memory B-cells already have the proper
antibodies stored up for that pathogen.
Immunology
An antigen
Immunity
that milkmaids who were exposed to cowpox
had immunity to smallpox
vaccination
“cow”.
cross-immunity
He worked with the bacteria that cause chicken
cholera. He accidentally found that old cultures
would not cause disease in chickens.
Subsequent injections of more virulent
organisms had no of effect on the chickens that
had been previously exposed to older cultures.
In this manner, the first attenuated vaccine was
discovered.
Change; may occur through heat, aging, or
chemicals
one that has been weakened, and it does not
cause disease.
Rabies
foreign objects introduced into transparent
starfish larvae became surrounded by white
blood cells that attempted to destroy the foreign
objects by eating them.
phagocytosis
direct cell-to-cell interaction
5
Immuno PPT Flashcards Unit 1
Humoral (“blood”) immunity involves
Who linked the two theories by showing that
the immune response involved both cellular and
humoral elements?
What did Wright observe?
The humoral, or circulating, factors are called
These serum factors include specific proteins
known as
These serum factors included also non-specific
factors called
The ability of the host to resist infection by
means of normally present body functions is
called
Is prior exposure required for innate immunity?
Does the response change with subsequent
exposures?
Innate immunity is the same as calling it
Characterized by specificity for each individual
pathogen, or microbial agent, and the ability to
remember a prior exposure, which results in an
increased immune response is
This refers to the action of the
Adaptive immunity is the same as calling it
Which immune responses are required for a
healthy host?
The two types of Adaptive Immunity are
Both types of immunities are acquired how?
The natural defense system can be considered as
being composed of which two parts?
Which system attempts to prevent entry of
pathogens?
What are the structural barriers that prevent
most infectious agents from entering the body?
Other external defenses include
(This includes monocytes/macrophages,
neutrophils, and Killer T-lymphocytes. It can
include eosinophils and basophils as well)
Antibodies
(This refers to the action of B-lymphocytes)
An English physician named Wright
that certain humoral, or circulating, factors
acted to coat bacteria so that they became more
susceptible to ingestion by phagocytic cells
opsonins
antibodies
acute-phase reactants
Innate immunity
No, they are nonspecific and are the same for
all pathogens or foreign substances to which
one is exposed.
The response does not change; the phagocytes
and Killer T cells are just doing their jobs by
identifying and eliminating pathogens that
might cause infection.
cellular immunity
Adaptive immunity
B-lymphocytes
humoral immunity
Both innate and adaptive
Active and Passive Immunity
Naturally and artificially
the external defense system and the internal
defense system
External system
unbroken skin and the mucosal membrane
surfaces
Lactic acid in sweat and fatty acids from
sebaceous glands to keep skin acidic and inhibit
6
Immuno PPT Flashcards Unit 1
What clear away 90% of the deposited material
in the respiratory tract?
What helps to remove many pathogens in the
renal system?
What plays role in preventing invasion of
pathogens in the female genital tract?
What halts bacterial growth in the digestive
tract?
Lysozyme is an enzyme found in tears and
saliva and it attacks the cell walls of bacteria,
especially Gram…?
Why is normal flora present on the body?
bacteria from growing.
mucous secretions and cilia
The flushing action of the urine, plus its slight
acidity
Lactic acid keeps the vagina acidic, preventing
invasion of pathogens
acidity of the stomach
-positive
to keep pathogens from establishing themselves
Which defense system deals with pathogens that The Internal defense system
gain entry?
What play essential parts in the internal defense Both cells and soluble factors
system?
The internal defense system is designed to
recognize molecules that are unique to
infectious organisms.
This typically involves recognizing what?
a carbohydrate such as mannose that is found in
bacteria and not on human cells
In internal defense system WBSs
seek out and destroy the invading cells
Phagocytosis is enhanced by soluble factors
acute-face reactants
called
Both internal and external defense systems
phagocytosis
promote what?
Acute phase reactants are produced primarily by hepatocytes (liver cells) within 12-24 hours in
response to cytokines
Chemicals that call other white blood cells to
Cytokines
the area of infection are called
Cytokines are produced by
monocytes and macrophages during the
inflammatory response
Cytokines are released, not only from infection, injury to the tissues
but also by
The main cytokines are:
Interleukin 1 beta (IL-1b), Interleukin -6 (IL-6)
and
Tumor necrosis Factor – alpha (TNF-a)
Acute phase reactants include
C-reactive protein
Serum amyloid A
The complement system
Mannose-binding protein
Alpha-1 antitrypsin
Haptoglobin
Fibrinogen
7
Immuno PPT Flashcards Unit 1
A substance found in small amounts in the
serum, thought to be an antibody to the
polysaccharide capsule of pneumococcal
bacteria is
When is CRP increases rapidly?
Within 48 hours after infection, the levels of
CRP can increase with how much?
How will the levels decline?
Elevated CRP levels are found in
CRP acts like an antibody and is capable of
CRP binds to specific receptors on
Because the levels of CRP rise and then decline
so rapidly, it is the most widely used indicator
of
CRP is a nonspecific indicator of disease or
trauma, but it can be used to
CRP is also used to monitor what?
How can CRP used to monitor malignancy and
organ transplantation rejection?
An increased level of CRP is also a significant
risk factor for
Another major protein whose concentration can
rise 1000 fold is
Where can Serum amyloid A found?
What is the role of Serum amyloid A?
By removing cholesterol from macrophages at
the site tissue injury, serum amyloid A
contributes to the
Serum amyloid A levels increases in what types
of infections?
A series of serum proteins that are normally
present and whose function is mediation of
inflammation refers to
How many complement proteins are there?
Complements are activated by
Ceruloplasmin
C-reactive protein (CRP)
after infection, surgery, or trauma
Levels can rise to 1000 fold
The levels decline rapidly when the stimulus
stops
bacterial infections, rheumatic fever, viral
infections, malignant diseases, tuberculosis, and
after a heart attack
optimization, agglutination, precipitation, and
activation of complement
monocytes, macrophages and neutrophils,
which promotes phagocytosis
acute inflammation
follow a disease process and observe the
response to treatment of inflammation and
infection
malignancy and organ transplantation rejection
A rise in its level may mean a return of
malignancy, or beginning of organ rejection
myocardial infarction and ischemic stroke in
people who have no previous history of
cardiovascular disease
Serum amyloid A
It is made in the liver and circulates in the
plasma
it is associated with HDL cholesterol, and is
thought to play a role in metabolism of
cholesterol
cleaning up of the area
more in bacterial infection than in viral
infections
Complement
9
antibodies
8
Immuno PPT Flashcards Unit 1
When are the complement proteins activated by
antibodies?
Several additional proteins are involved in the
The alternate pathway is triggered by
The major functions of complement are
Which protein is able to recognize foreign
carbohydrates such as mannose which are found
on bacteria, yeast, viruses, and parasites?
How does MBP acts?
Where can MBP found?
What happens when MBP binds to the sugar?
in an event called classical complement cascade
alternate pathway
microorganisms
opsinization, chemotaxis, and lysis of cells
Mannose-binding protein (MBP)
MBP acts as an opsonin
MBP is widely distributed on mucosal surfaces
throughout the body
It activates the complement cascade and helps
to promote phagocytosis
recurrent yeast infections
Alpha-1 antitrypsin (AAT)
Lack of MBP is associated with
A general Plasma inhibitor of proteases released
from leukocytes, especially elastase, which is an
enzyme that degrades elastin and collagen is
called
Because of its activity in chronic pulmonary
Lung tissue
disease, what is damaged?
During an inflammatory response, what does
It counteracts the effects of neutrophil invasion
AAT do?
AAT also regulates what?
Expression of cytokines such as TNF-a, IL-b,
and IL-6.
AAT deficiency can result in
premature emphysema in smokers since
uninhibited proteases remain in the lower
respiratory tract, leading to destruction of cells
in the lungs
AAT can also react with
any serine protease, such as those generated by
the complement cascade
Once bound to AAT, what happens to the
the protease is completely inactivated
protease?
Haptoglobin’s job it to bind to what that is
free hemoglobin
released when RBCs are lysed i
Once haptoglobin is bound, the complex is
Kupffer cells in the liver
ingested rapidly by
Why are the Kupffer cells ingest the complex?
Once bound, the complex is ingested rapidly by
Kupffer cells in the liver so the kidneys are not
damaged by trying to filter out that large protein
What do the Kupfer cells prevent?
the loss of iron
Where is the complex sent?
it can be sent to the bone marrow to be recycled
Haptoglobin also protects against
oxidative damage that the free hemoglobin
would cause
The rise of haptoglobin in the plasma is due to
liver making too much
the
What does the rise of haptoglobin indicates?
inflammation, stress, or tissue necrosis
9
Immuno PPT Flashcards Unit 1
Early on in the inflammatory response,
haptoglobin levels may drop because of
What levels must be evaluated along with the
other acute phase reactants?
Which one is the most abundant of the
coagulation factors in plasma?
What does fibrinogen do?
Why is the formation of fibrin blood clot
important?
Fibrinogen also serves to promote
Increased levels of the fibrinogen contribute to
The main copper-transporting protein in human
plasma is
How is Ceruloplasmin oxidizes iron?
Fe3+ releases iron from ferritin, so it can bind
to what?
What happen to RBC’s that are old?
After the RBC’s are trapped, what happen to
them?
Hemoglobin is made from
These three segments of hemoglobin are
The globin chains (proteins) are broken down
into
Amino acids are used for
In the hemoglobin what are the globins?
The heme units are the
What is essential to nearly all cells but also is
quite toxic?
Iron is essential for its role in
intravascular hemolysis; consequently masking
the protein’s behaviour as an acute phase
reactant
Plasma levels
Fibrinogen
it forms the fibrin blood clot
A clot stops bleeding and also creates a barrier
that helps prevent the spread of microorganisms
further into the body
aggregation of RBCs
an increased risk for developing coronary artery
disease, especially in women
Ceruloplasmin
from Fe2+ to Fe3+, which releases iron from
ferritin
transferrin
their cell membranes become irregular and they
become trapped in the reticular fibers in the
spleen
A macrophage engulfs it, and breaks down the
hemoglobin
heme (a porphyrrin ring with iron in the center),
and globin chains
detached from each other and liberated
amino acids (the building blocks of proteins)
synthesis of any other proteins wherever they
are needed
The alpha and beta chains
the porphyrin rings
Iron
oxidation-reduction reactions needed for
metabolism
Where is iron’s toxicity comes from?
from its propensity to form oxygen radicals that
damage cells.
What happen when hemoglobin is broken
the iron (Fe+2) is released into plasma
down?
(ceruloplasmin is needed for that).
Which protein binds to iron?
apoferrin
After apoferrin binds to the iron, the apoferrin is transferrin
called
What does transferring do?
takes the iron into cells that can use or store it
10
Immuno PPT Flashcards Unit 1
The iron is stored in most body cells as
A depletion of ceruloplasmin is found in what
disease?
Wilson’s disease, which is characterized by
Copper accumulates where in the body?
Normally, the circulating copper is found
where?
Cellular defense mechanisms include which 6
main types of WBCs?
About how many percent of all WBCs are
basophils?
How big are the basophils when comparing
them to the other granulocytes?
The granules of the basophils contain
What are histamine, heparin, and eosinophilic
chemotactic factor-A do?
Histamine causes what?
Vasodilation of the blood vessels allows
What is Heparin?
Which immunoglobin is formed in allergic
reactions?
What is the role of IgE?
What happens when IgE binds to basophil cell
membranes?
What is a Mast Cell?
What is the difference between the mast cells
and basophils?
Mast cells live how long comparing to
basophils?
The enzyme of the Mast cells’ granules contain
What is the function of mast cells?
About how many percent of all WBCs are
eosinophils?
When is the number of eosinophils increased?
ferritin (Fe+3) or as hemosiderin in white blood
cells.
Wilson’s disease
massive increase of copper in the tissues
in the brain, cornea, kidneys, and bones
It is absorbed by the liver and combined with
ceruloplasmin and returned to the plasma
Basophils (in blood vessels) and Mast Cells (in
tissues)
Eosinophils
Neutrophils
Dendritic Cells
Monocytes
Natural Killer Cells
Less than 1%
They are the smallest of the granulocytes
histamine, heparin, and eosinophilic
chemotactic factor-A
induce immediate hypersensitivity reactions
vasodilation of the blood vessels
white blood cells to migrate to the tissues faster,
but also allows the plasma to leak out, which
causes stuffiness and discomfort
an anticoagulant
IgE
IgE binds to basophil cell membranes
causing the granules to release when they
contact an antigen
A mast cell is similar to a basophil, but exists in
the tissues
The mast cells are larger and have more
granules than basophils
They live 9-18 months, whereas basophils only
live a few days.
acid phosphatase, alkaline phosphatase, and
protease
It functions the same as a basophil, by playing a
role in hypersensitivity reactions by binding IgE
1-3%
during an allergic reaction or in response to a
11
Immuno PPT Flashcards Unit 1
The eosinophil’s granules contain
The eosinophil-specific granules contain
What are the eosinophils capable of?
How efficient the eosinophils are?
The most important role of the eosinophils is
Another name for Neutrophils are also called?
Why are neutrophils sometime called “segs”?
How many granules does a neutrophils contain?
What color is primary granules contain?
What defensive properties does primary
granules contain?
What does secondary granules have?
What does tertiary granules contain?
Acid hydrolases are called what, which are
found in separate compartments?
About what percentage of neutrophils are found
in the blood vessels?
Neutrophil circulate in the blood for how long?
When WBC enter blood vessel tissue by a
process called what?
What the name of the receptor that make
neutrophils become stick y like Velcro against
the inside wall of the blood vessel?
Diapedesis occurs when neutrophil sense a
what?
When neutrophils move toward or away from a
chemical is called what?
What the chemical messengers that cause cells
to migrate in a particular direction?
Some of the factor that effect chemotactic for
neurtophils included?
What the life span of a neutrophil once in the
tissue?
Named because they care covered with long
membranous extensions that make them
parasitic infection
acid phosphatase
several different proteins including peroxidase
and neurotoxin
They are capable of phagocytosis
not efficient because they lack digestive
enzymes
neutralizing basophil and mast cell products and
killing certain parasites
Polymorphonuclear Neutrophils (PMN’s)
Because of their segmented nucleus
3 types of granules
-Primary granules
-Secondary granules
-Tertiary granules
Blue
Antibacterial proteins
-collagenase, lactoferrin, lysozyme, NADPH,
and protein found in cell membranes.
Gelatinase and plasminogen activator
Lysosomes
About 50%
About 6-10 hours
Diapedesis
Selectins
Cytokine
Chemotaxis
Chemotaxins
-complement components
-proteins from coagulation
-product of virus and bacteria
-platlet activating factor
-secretion from mask cells
5 days
Dendritic cells
12
Immuno PPT Flashcards Unit 1
resemble nerve cell?
What the main function of dentritic cells?
Dendritic cells are classified according to their
tissue location, true or false?
Dendritic cells found on the skin and mucous
membranes are called what?
Dendritic cells found in the heart, lungs, liver,
kidney, GI tracts are called what?
In the secondary lymphoid tissue and thymus
they are also called what?
After dendritic cells capture an antigen by
phagocytosis or endocytosis, where do they
migrate?
Dendritic cells are the most potent phagocytic
cell in the tissue, true or false?
What the name for monocytes when found in
the tissue
What he larges blood cells?
What the shape of the monocyte?
What the appearance of the monocytes?
Phagocytose antigen and present it to helper T
Lymphocytes
True
Langerhans cells
Interstitial dendritic cells
Interdigitating dentrici cells
To the blood and lymph organs where they
present antigen to T-lymphocytes to initiate the
adaptive (humoral) immune response of making
antibodies
True
macrophages
Monocytes
“horseshoe”shaped
Grayish blue and has a ground glass appearance
due to presence of fine dust-like granules
Peroxidase, acid phosphatase, and arylsulfatase
One type of monocyte contain similar property
to neutrophils such as?
Other type of monocyte granules contains what? Glucuronidase, lysozyme, and lipse, but no
alkaline phosphatase.
What percentage does monocyte make up of
About 5-10%
circulating WBCs?
How long do monocyte stay in circulation?
About 3 days
After circulation for 3 days where they migrate To the tissue, where they are now called
to?
macrophages
Once monocytes enter the tissue, it is now
Macrophage, becomes larger and lose its
called what?
peroxidase
Lung macrophages are called what?
Alveolar macrophages
In the liver they are called what?
Kupffer cells
In the brain they are called what?
Microglial cells
In the connective tissue they are called what?
histiocytes
When compare to neutrophil, are monocytes
Slower, they are not as efficient as neutrophil in
faster or slower?
phagocytosis
When compare to neutrophil do monocytes live Longer, can live for months instead of days
longer or shorter?
What role do monocyte perform?
Help initiating and regulating the immune
response, microbial killing, turmoricidal
activity, intracellular parasite eradication,
secretion of cell mediators, and antigen
13
Immuno PPT Flashcards Unit 1
Killing activity is enhanced when they become
activated by what?
What cell are the only lymphocytes that
participate in cellular (innate) immunity?
What can natural killer cells do?
What the mechanism of natural killer cells?
Does Natural killer cell participate in
phagocytosis?
Additional set of mechanism on certain cells are
what?
What exactly is Toll?
Similarity of toll-like receptors are also found
where?
Higher concentration of toll-like receptors
occurs on what WBCs?
Each of toll-like receptors recognizes a different
microbial product, true or false?
How does toll-like receptors function?
What kind of immunity does toll-like receptors
enhance?
How many steps occurs in phagocytosis?
What are the 4 main steps?
presentation.
Contact with microorganism or cytokines which
are release by T-lymphocytes
Natural killer cells
They are able to recognize self from non-self
materials
They will pop cell membrane of microorganis
No
Toll-like receptors
It is a protein originally discovered in the frut
fly Drosophila, where it plays an important role
in antifungal immunity in adult
Similar molecules are found on human
leukocytes and some non-leukocyte cell types
Monocytes, macrophages, and neutrophils
True
Once a receptor binds to its particular
substance, or ligand, phagocytosis may be
stimulated, or the cell produces cytokins that
enhance inflammation and eventual destruction
of the microorganism.
Innate immunity
4 main steps
1. Physical contact between the white cell
and the foreign particle
2. Formation of a phagosome
3. Fusion with cytoplasmic granules to
form a phagolysosome
4. Digestion and release of debris to the
outside
Chemotaxis from chemicals such as bacterial
factors, complement components, or CRP
Enhanced by opsonins
Neutrophils and macrophages migrate by what
mechanism?
Receptors on the phagocyte come in contact
with foreign particle surface and activates what?
What are opsonins?
They are serum proteins that attach to the
foreign substance, such as antibodies, CRP, and
complement components.
How does phagocytosis work?
Phagocyte has receptors that attach to the
opsonin, and grab it like a handle so it can
ingest the foreign object or bacterium
14
Immuno PPT Flashcards Unit 1
How does opsonin help in phagocytosis?
What is opsonization?
Opsonization are perform by opsonin serum
protein, true or false?
What the process of phagocytosis?
An increase in oxygen consumption in
phagocytosis is termed
Once the particle is inside the phagocyte the
vacuole is called what?
When the cytoplasmic granule fuse to the
phagosome, and the entire complex is now
called what?
The granules release their contents, and
digestion occurs, true or false?
Any undigested material is excreted by what?
The duration of the whole process of
phagocytosis take how long?
During the respiratory burst or oxidative burst,
the bacterial activates what?
Superoxide is highly toxic but can be rapidly
converted to more lethal products, true or false?
By adding hydrogen ions, the enzyme
superoxide disumatase converts superoxide to
what?
Hypochlorite ions help potentiated the effect,
true or false?
How is this accomplished?
New evidence indicates that an enzyme known
as what, will depolarize the bacterial cell
membrane, allowing hydrogen potassium ions
to enter the vacuole?
When hydrogen combines with the superoxide,
what happen to the pH? Increase or decrease?
What does increase in pH activates?
The dysfunction of the NADPH oxidase system
causes what disease?
What the out come of chronic granulomatous
Opsonin may act by neutralizing the surface
charge on the foreign particle, making it easier
for the cells to approach one another
Is the process whereby a substance binds to the
foreign object to enhance phagocytosis
True
Once attachment occurs, the phagocytes’
cytoplasm forms pseudopods and engulfs the
particle. This cause an increase in oxygen
consumption.
Respiratory burst or oxidative burst
Phagosome
Phagolysosomes
True
Exocytosis
About 20 seconds
The cell membrane of the phagocytic cell, and a
radical known superoxide is formed
True
To hydrogen peroxide or the hydroxyl racial
OH
True
Action of the enzyme myeloperoxidase in the
presence of chloride ions. Hypochlorite ions are
powerful oxidizing agents.
Enzyme from Electron transport system called
NAPDH
Increase pH
Activates protease (protein digesting enzymes)
which contribute to microbe killing
Chronic granulomatous disease
Patients suffer from recurring, severe bacterial
15
Immuno PPT Flashcards Unit 1
disease?
What is termed inflammation?
What immunity are involved?
What are the four clinical symptoms of
inflammations?
What are some examples of major events that
associated with process of inflammation?
When inflammation occurs what chemical
mediator are release?
What the purpose of histamine?
Vasodilation allows what to leaked into the
tissues?
When vasodilation causes plasma to leaked in to
the tissue causes what to happen?
What soluble mediators initiate and control the
response?
Amplification occurs through what formation?
As the endothelial cells contract, which WBCs
move out of circulation and into the tissue?
Neutrophils are attracted by what substance?
Neutrophils are mobilized within how many
minutes after the injury?
How long does neutrophil emigration last?
Migratin of macrophates from surrounding
tissue and from blood monocytes occurs when?
Macrophages attempt to clear the area through
what process?
When the inflammation process becomes
proglonged it is said to be what?
What may contribute to the damage?
Failure to remove microorganism or injured
infections
Overall reaction of the body to injury or
invasion by an infectious agent.
Both cellular and humoral mechanism are
involved
• Redness (rubor)
• Swelling (tumor)
• Heat (calor)
• Pain (dolor)
• Increased blood supply to the infected
area
• Increased capillary permeability
• Migration of white blood cells, mainly
neutrophils to the injured area
• Migration of macrophages to the injured
area
Histamine
Released from injured mast cells which causes
dilation of blood vessels and bring additional
blood flow to the affected area, resulting in
redness and heat
Plasma
Produce swelling, and pain from the fluid
pressing on nerves
Acute-phaase reactants
Formation of clots by the coagulation system
and then the triggering of the fibrinolytic
system
Neutrophils
Chemotaxis
30-60 minutes
24-48 hours
Several hours later and peaked at 16 to 48 hours
Phagocytosis
Chronic, and tissue damage and loss of function
may result
Specific immunity with infiltration of
lymphocytes
Continued tissue damage
16
Immuno PPT Flashcards Unit 1
tissue may results in what?
What is the most widely monitored of the acute
phase reactants and is the best indicator of acute
inflammation?
What are of primary importance in the
lymphoid system’s immune response?
Lymphocytes represent between what percent of
the circulating white blood cell?
The size of small lymphocyte is typically
between what in diameter?
Does a lymphocyte have a nucleus?
What color does the nucleus stain?
What does the lympocyte’s cytoplasm
container?
What is lymphocytes rise from?
Where is hematopoietic stem cell further
differentiate in?
Where is the primary lymphoid organs in
human located?
The lymphocytes that go from the bone marrow
to the thymus glad differentiate into what?
Those that stay in the bone marrow to
differentiate are called what?
The secondary organs in the lymphoid system
includes what?
The main contanct with foreign antigens take
place in secondary or primary organ?
The spleen is part of what lymphoid system?
The spleen serve what purpose?
What does the lymph nodes do?
Mucosal surface are equip with what?
The circulation of lymphatic fluid are controlled
by what?
T lymphocytes are also known as what type of
cell that serve a regulatory role?
What the main purpose of B lymphocytes
Which lymphocytes recirculate continuously
from the bloodstream to the secondary
lymphoid organs and back, to increase contact
with foreign antigens?
A third type of lymphocyte, with large granules,
plays a role in both the innate and adaptive
immune response?
C-reactive protein (CRP)
Lymphocytes
About 20-40%
between 7 and 10 μm
Yes
Deep blue
Contains a few organelles with no specific
granules, and stains a lighter blue
Hematopoietic stem cell in the bone marrow
Primary lymphoid organs
Bone marrow and the thymus
T-cells (T for thymus)
B-cell (bone marrow)
Spleen, lymph nodes, appendix,tonsils, and
other mucosal-associated lymphoid tissue
Secondary organs
Secondary organ
Serves as a filtering mechanism for antigens in
the bloodstream
Help filter fluid from the tissue
Backed with lymphoid tissue as an additional
means of contacting foreign antigens as they
enter the body
cytokines
Effector cells
Produce antibodies
Both T and B lymphocytes
NK cells (Natural Killer cell)
17
Immuno PPT Flashcards Unit 1
T,B, and NK cell arise from what common
precursor?
In the peripheral blood, what percent of all
lymphocyte make up B cells?
In the peripheral blood, what percent of all
lymphocyte make up T cells?
In the peripheral blood, what percent of all
lymphocyte make up NK cells?
What are the two primary lymphoid organs?
All lymphocytes arise from what stem cell?
Where does pluripotential hematopoietic stem
cells found in?
The bone marrow assumes this role when the
infant is born, true or false?
What organ can be considered the largest tissue
of the body?
Where are bone marrow found?
Which bone marrow is the source of the
hematopoietic stem cells?
What can red bone marrow give rise to?
The bone marrow functions as what?
T, B, and NK cells arise from what precursor?
Lymphocyte leave what organ and migrate to
the primary lymphoid organs where they
differentiate further?
Some of these cells will go to the thymus and
develop into what cell?
While the other remain in the bone marrow and
mature into what?
T cells develop their identity characteristic in
what organ?
Where is the thymus organ located?
The thymus gradually atrophies at what age?
Most of the virgin T cells are produce during
when?
The thymus are capable of producing new T
cells until what age?
Progenitors of T cells appear in the fetus as
early as?
By the time the infant is born the differentiation
of lymphocytes take place during when?
There are two different layer of the thymus
Common lymphoid precursor (CLP)
About 10-20%
About 61-89%
Up to 22%
Bone marrow and Thymus
Pluripotential hematopoietic stem cell
Yolk sac of developing embryo and are later
found in the fetal liver
True
Bone marrow
In the epiphyses of long bones and is also found
in flat bones of the body, especially the iliac
crest, the vertebral bodies, and the sternum
Red bone marrow
Erythrocytes, granulocytes, monocytes,
platelets, and lymphocytes
The center for antigen-independent
lymphopoiesis
Common lymphoid precursor (CLP)
Red bone marrow
T cells
B cells
Thymus
Sits on top of the heart
After puberty
First three years of life
Fifth or sixth decade of life
Eight weeks in utero
Early in fetal development
Cortex (outer edge)
18
Immuno PPT Flashcards Unit 1
called?
Mature T lymphocytes are released from what
area of the thymus?
Once differentiation occurs, mature T
lymphocytes are released from what organ?
Once differentiation occurs, mature B
lymphocytes are released from what organ?
After differentiation is complete where do
lymphocytes migrate to?
Secondary lymphoid organs consist of what
tissue?
The lymphocytes travel through the tissue and
return to the bloodstream by what body part?
Each lymphocytes spend most of its life in what
kind of tissue?
Lymphocytes go from one secondary organ to
another via what route?
Recirculation increase the likelihood of a
lymphocyte to come in contact with what?
Lymphopoiesis occurs in what tissue?
The reproduction of lymphocytes is termed
what?
Most naïve or resting lymphocytes die with in
how many days?
Within a few days most lymphocyte will die if
they are not activated by the presence of what?
It is this second process that gives rise to the
long-lived?
What is the largest secondary lymphoid organ?
Where is the spleen located?
What the purpose of the spleen?
The spllen has two type of tissue known as
what?
What percentage does the red pulp make up?
What is the function of red pulp?
Blood flow into the red pulp via what?
Blood flow from the arterioles into the red pulp
but exits through what vein?
What percentage does the white pulp make up?
The sheath within the white pulp contain what
cells?
Medulla (inner area)
Medulla (inner area)
Thymus
Bone marrow
Secondary lymphoid organs and become part of
the the recirculating pool
Spleen,lymph nodes, tonsils, appendix, peyer’s
patches in the intestines, and other mucosalassociated lymphoid tissue
Thoracic duct
Solid tissue
Circulation
Specific antigen
Secondary tissue
Lymphopoiesis
Within a few days
Foreign antigen
Memory cells
Spleen
Upper left quadrant of the abdomen just below
the diaphragm and surround by a connective
tissue capsule
Act like a sponge to help remove old and
damaged cells and foreign antigens from the
blood
Red pulp and white pulp
More than one-half of the spleen
Function to destroy old red blood cells
Arterioles
Splenic vein
About 20% of the spleen
T cells
19
Immuno PPT Flashcards Unit 1
Attached to this sheath are primary follicles,
which also contain what cell that are not yet
stimulated by antigen?
Surrouding the PALS (periarteriolar lymphoid
sheath) containing what cells?
Lymphocytes enter and leave the spleen by
mean of many what?
Each day blood volume passes through the
spleen approximately how many times?
At the same time allow what WBCs to
constantly survey for infectious agents and
other foreign matter?
The white pulp composes of approximately
what percentage of the total weight of the
spleen?
White pulp contains what kind of tissue?
Where are lymph nodes located?
What is the purpose of lymph nodes?
Lymph fluid arises from what?
Some of the interstitial fluid returns to the
bloodstream via what?
Lymph nodes are especially numerous near
what body part?
Filtration is a main function of what tissue?
The lymph nodes contains what, which are lined
with macrophages?
The lymph node is organized into how many
parts?
The lymph fluid flows slowly through spaces
called what?
Sinuses are line with what?
Macrophages allow what process to take place?
Foreign antigens enter the lymph nodes through
what vessels?
Numerous lymphocytes also enter the nodes
from what?
The outermost layer of the lymph node is called
what ?
The cortex of the lymph nodes contains what?
Specialized cells called what are also located in
the primary follicles?
B cells
Dendritic cells that trap antigen
Capillary branches that connect to the arterioles
Approximately 4 times
Lymphocytes and macrophages
20%
Lymphoid tissue
Along lymphatic ducts
Serve as central collecting points for lymph
fluid from adjacent tissue
Passage of fluids and low-molecular-weight
solutes out of the blood vessel walls and into
the interstitial spaces between cells
Venules
Joints and where the arm and legs join the body
Lymph nodes
Sinuses
Outer cortex
Paracortex
Medulla
Sinuses
Macrophages
Phagocytosis
Afferent lymphatic vessel
Bloodstream by mean of specialized venules
called high endothelial venules
The cortex
Macrophages and aggregations of B cells in
primary follicles
Follicular dendritic cells
20
Immuno PPT Flashcards Unit 1
Follicular dendritic cells are found only in
what?
Follicular dendritic cells exhibit large number
of what?
The secondary follicles consist of what?
The interior of a secondary follicle is known as
what?
What so important about germinal center?
What does plasma cell secrete?
B-cell memory is a primary function of?
Where does T lymphocytes mainly localized?
Paracortex is located where?
The medulla is more or less dense?
Medulla contains what cells?
Particulate antigens are removed as the fluid
travel across what?
The transit time through a lymph node is about
how many hours?
Fluid and lymphocytes exit by way of what?
Lymph nodes eventually connect with the
thoracic duct and the venous system to
recirculate what?
Additional areas of lymphoid tissues includes?
MALT is found where?
What does Peyer’s patches represent?
Another area of lymphoid tissue found in the
mucus membrane lining of the oral and
pharyngeal cavities
Additional location of lymphoid tissue is the
what?
All of these secondary organ function as what?
The cutaneous-associated lymphoid tissue with
the epidermis contains what?
Within each of these secondary organs, which
cells are segregated and perform specialized
functions?
B cells differentiate into what?
Plasma cell and memory cells are responsible
Lymphoid follicles and have long cytoplasmic
processes that radiate out like tenticles
Receptors for antibody and complement and
help to capture antigen to present it to T cells
and B cells
Antigen-stimulated proliferating B cells
Germinal center
It is where blast transformation of B cells take
place
Secrete antibody and memory cells
Lymph nodes
Paracortex
Region between the follicles and the medulla
Less dense
T cells in addition to B cells, macrophages, and
numerous plasma cells
Lymph node from cortex to medulla
18 hours
Efferent lymph vessels
Between lymph nodes and blood
MALT, tonsils, appendix, and cutaneousassociated lymphoid tissue
In the gastrointestinal, respirator, and urogenital
tracts
Represent a specialized type of MALT and are
located at the lower ileum of the intestinal tract
tonsils
Appendix
Potential sites for contact with foreign antigen,
and they increase the probability of an immune
response.
Intraepidermal lymphocytes, where most of
these are T cells
T and B cells
Plasma cells and memory cells
Humoral immunity or antibody formation
21
Immuno PPT Flashcards Unit 1
for what immunity?
T cells play a role in what immunity?
B cells are derived from what precursor?
Common lymphoid precursor differentiate into
what?
Early lymphocyte progenitors can becomes
what?
What are some example of differentiation of B
cell?
The earliest B-cell precursor can be recognized
by the presence of a surface molecule called?
What so important about antigen-independent
phase?
How does influence of growth factors and
cytokines effect the differentiation of B cells?
After the rearrangement of genes occurs what is
the outcome?
When B lymphocytes produce two identical
light chains and two identical heavy chains,
what is this process called?
What are some example of markers that exist on
surface of pro-B cells?
In order for pro-B cells to differentiate into preB cell successfully what must occur?
What cell interact with pro-B cells to secrete
cytokines, hormones, chemokines, and adhesion
molecules?
What does the pre-B stage begins?
After pre-B cells is complete which marker
surface marker does it lose?
Where does Mu chains accumulate?
Which combination of chain forms the pre-B
cell receptors?
The pre-B cell receptor adheres to what part of
Cell-mediated immunity and produce sensitized
lymphocytes that secrete cytokines
Common lymphoid precursor (CLP)
B cell progenitor cell
T-cell, B-cell NK cells, or dendritic cells
•
•
•
•
•
•
•
Pro- B cells
Pre-B cells
Immature B cells
Mature B cells
Activated B cells
Plasma cells
Memory cells
CD45R
Process that prepares them for their role in
antibody production and restricts the types of
antigen to which the cell can respond.
This causes rearrangement of genes to occur
and codes for the heavy and light chains of an
antibody molecule.
B lymphocyte programmed to produce a unique
antibody molecule, consisting of two identical
light chains and two identical heavy chains.
Pro-B cell
CD19, CD45R, CD43, CD24, and c-Kit
Successful rearrangement of heavy-chains
genes.
Stromal cells
When synthesis of the heavy chain part of the
antibody molecule occurs
CD43 marker as well as c-Kit and TdT
In the cytoplasm
Combination of two heavy chains and two
surrogate light chains plus two very short (Ig-α
and Ig-β)
Bone marrow stromal cell membranes and
22
Immuno PPT Flashcards Unit 1
the body?
Which cell only express heavy chains in
association with surrogate light chain?
The next stage after pre-B cells is known as
what?
Immature B cells have what molecules on the
cell surface?
Its specificity is determine by what?
What other surface protein appears on the
immature B cell?
The receptor that help breakdown product of the
complement component C3, and C3d?
Antigen coated with complement fragments
during the immune response help trigger what?
CD40 and MHC class II are important for
which interaction?
Many B cells are deleted form the marrow by
the process of programmed cell death, termed?
Which organs doe immature B cells leave and
proceed to?
In the spleen, what does immature B cells
develop into?
Mature B –cells in the spleen is also called
what?
B cells remain in the spleen to do what?
Other immature B cells differentiate into what
and where are they found?
Besides IgM antibody, which class of antibody
are exhibit?
How does IgD affect the life span of mature B
cells in the periphery
Unless contact with antigen occurs, the life span
of a mature B cell is typically how many days?
What occurs when B cell is stimulated by
antigen?
The transformation process is known as what?
What the half-life of B cells?
Where does antigen-dependent activation of B
cells take place in?
Activated B cells exhibit identify markers
including?
What is Interleukin- 2?
transmits a signal to prevent rearrangement of
any heavy-chains genes
Pre-B cells
Immature B cell
IgM molecules
Variable regions, which occur on both the light
and heavy chains of the antibody molecules
CD21, CD40, and MHC class II molecules
CD21
Enhances the likelihood of contact between B
cells and antigen
B cells with T cells
Apoptosis
Leave bone marrow and proceed to seed the
spleen and other secondary lymphoid organs
Mature B- cells
Marginal zone B cells
Respond quickly to any bloodborne pathogens
they may come in contact with
Become follicular B cells, which are found in
the lymph nodes and other secondary organs
All mature B cells exhibit IgD on the surface
Prolong it
Few days
It undergoes transformation to a blast stage,
which eventually forms memory cells and
antibody-secreting plasma cells
Antigen-dependent phase of B cell development
More than 6 weeks
The primary follicles of peripheral lymphoid
tissue
CD25, which is found on both activated T and
B cells and acts as a receptor for Interleukin 2.
A growth factor produced by T cells
23
Immuno PPT Flashcards Unit 1
When B cells are activated in this manner, they
transform into what?
What are the size of plasma cells?
Does a plasma cell have abundant cytoplasmic
immunoglobulin or abundant surface
immunoglobulin
Where are plasma cells located?
Do plasma cells divide?
Plasma cells can differentiate into what type of
cells?
Where are memory B cells located?
How long due memory cells remain activated?
What percentage of circulating lymphocytes are
T cells?
What are lymphocyte precursors called?
Where do thymocytes enter and migrate?
What is T cell differentiation driven by?
What are the early surface markers of T cells?
Are distinct surface markers created during
specific stages of development
How long does T cell differentiation take?
What are the thymic stromal cells?
What percentage of the cortical cells die
intrathymically before maturing completely?
Early thymocytes lack which CD markers?
Double negative thymocytes proliferate under
the influence of which IL?
How many chains does CD3 have?
The alpha and beta chains have variable
regions. What does that mean?
When a functional beta chain is seen on the cell
surface, what happens?
Signaling by the beta chain triggers what
response in the thymocyte?
Do thymocytesexpress gamma and delta chains?
Thymocytes with gamma and delta chains are +
or – for CD4 and CD8?
Where are negative CD4 and negative CD8
cells located?
Cells that are negative for CD4 and negative for
CD8 may behave like what other type of cell?
When cells express both CD4 and CD8
antigens, what are the cells called?
Blast cells that will give rise to both plasma
cells and so-called memory cells
10-20um
Abundant cytoplasmic immunoglobulin
In the germinal centers of peripheral lymphoid
organs
No
Memory B cells
Germinal centers
Months or years
60-80%
Thymocytes
They enter the thymus and migrate to the cortex
Chemokines
CD44 and CD25
Yes
3 weeks
Epithelial cells, macrophages, fibroblasts and
dendritic cells
97%
CD4 and CD8
IL-7
8
It means that those regions recognize specific
antigens
A signal is sent to suppress further beta chain
gene rearrangements
Allows thymocyte to become CD4 and CD8
positive
Yes
Negative
Intestinal epithelium and pulmonary epithelium
Natural Killer cell
Double positive cells
24
Immuno PPT Flashcards Unit 1
Thymocytes that are unable to recognize self
antigens die where?
Approximately how many double positive
thymocytes survive T cell differentiation?
Thymocytes that express both CD4 and CD8
antigens are called what?
True/False: Survivors of selection exhibit only 1
type of marker, either CD4 or CD8.
CD4+ cells are also known by what name?
CD8+ cells are also known by what name?
What MHC class do CD8+ cells recognize?
What lymphoid organ are mature T cells
released from?
In peripheral organs, how long can resting T
cells live?
What do TH1 cells produce?
Which interleukins do the Th2 cells produce?
What is the main role of the TH 2 cell?
Which antigens do T-regs possess?
What is the main function of T regs?
How do T regs suppress the immune response?
What are the 3 types of T cells?
What CD markers do NK cells have?
What CD markers do helper T cells have?
Single positive T cells spend how many days in
the thymic medulla?
What do T regulatory cells prevent?
After leaving the thymus, where do T cells
migrate?
Where must antigens be transported for
activation?
T lymphocytes transform into what cells when
antigen recognition occurs?
Activated T lymphocytes express receptors for
what IL?
What do T lymphoblasts differentiate into?
Do memory T cells express a broader or limited
array of cytokines?
All activities of T cells make up what?
Do natural killer cells express the markers of T
In the thymus
1-3%
Double positive
True
T-helper or inducer cells
T-cytotoxic cells
MHC class I
Thymus
Several years
IFN-B and INF-Y
IL-4, IL05, IL-10, and IL-13
To help B cells produce antibodies
CD4 and CD25
To suppress the immune response to selfantigens
They produce interleukins such as IL-10 and
transforming growth factor B
Helper T cells, Natural killer cells, and
suppressor T cells
CD8
CD4
12 days
They prevent autoimmune reactions with any
surviving T cells that can react with self
antigens
Secondary lymphoid organ
T cell zones of the secondary lymphoid tissue
Large activated cells
IL-2
Functionally active small lymphocytes
Broader
Cell mediated immunity
No/Neither
25
Immuno PPT Flashcards Unit 1
or B cells?
What percentage of the circulating lymphoid
pool do natural killer cells make up?
What is a unique ability of the natural killer
cell?
True/False: Natural killer cells lack specificity
in their response?
What cell line do NK cells arise from?
NK cells in the bone marrow respond to which
IL to become NK cells?
NK cells present in the thymus become what
cell?
What are the 2 CD markers present on NK
cells?
NK cells high in CD56 and with little CD16
have what function?
NK cells high in CD16 but low in CD56
provide what function?
Name the 3 cytokines that stimulate NK cells.
Healthy self cells have which MHC class
proteins on their membrane?
What substances do NK cells release to kill
cells?
What is the function of granzymes?
What laboratory method is used to identify T
and B lymphocytes?
What are the 5 CD markers recognized on T
cells?
What are the 3 CD markers recognized on B
cells?
What is an antigen?
What 3 factors cause a lack of immune
response?
Name the 5 factors that influence the nature of
the immune response.
What 3 populations have decreased immune
responses?
True/False: The larger the dose of an
immunogen one is exposed = greater the
immune response
What happens if a person receives large doses
of an immunogen?
Does the amount of immunogen needed to
5-10%
To kill target cells without prior exposure to
them
True
Common lymphocyte precursor
IL-15
T cells
CD 16 and CD 56
Produce cytokines to support antibody
production
Produce cytokines to support cytotoxic activity
Il-12, interferon gamma, and interferon beta
MHC I
Perforins and granzymes
To lyse the cell
Flow cytometry
CD2, 3, 4, 7, and 8
CD19, 20, and 22
A substance that reacts with an antibody or
sensitized T cell, but may not invoke an
immune response
Nature of the immunogen, genetic coding of
MHC molecules, immunogen processing and
presentation
Age, overall health, dose, route of transmission,
and genetic capacity
Older people, newborns, and people in poor
health
True
T and B cell tolerance
Yes
26
Immuno PPT Flashcards Unit 1
generate an immune response differ with route
of inoculation?
Name the 4 routes for an immunogen
Does genetic predisposition play a role in
individual’s response to a particular
immunogen?
Name the 4 characteristics that influence the
ability of an immunogen to stimulate a host
What is the minimum molecular weight of an
immunogen to be recognized by the immune
system?
The _______ the molecular weight, the more
potent the molecule is an immunogen
What are the 2 best immunogens?
What are the proteins composed of?
If a string of amino acids are in a helix, is that
primary, secondary, or tertiary structure?
What do proteins contain that stimulate T cells?
Are synthetic polymers immunogenic?
Since synthetic polymers lack immunogenicity,
what are they used to make?
Why are carbohydrates less immunogenic than
proteins?
As immunogens, carbohydrates occur most
often in what form?
Are the A and B blood antigens glycolipids or
glycoproteins?
Are the Rh antigens glycolipids or
glycoproteins?
Substances recognized by the immune system
as non-self are immunogenic or nonimmunogenic?
What happens to lymphocytes that are capable
of reacting with self antigens?
What is a better immunogen, a plant or animal
protein?
True/False: Pure nucleic acids and lipids are
immunogenic by themselves
Is foreignness a characteristic that all
immunogens share?
What is antigen processing?
For a substance to elicit an immune response,
IV, intradermal, subcutaneous, and oral
Yes
Macromolecular size, chemical composition
and molecular complexity, foreignness, ability
to be processed and presented with MHC
molecules
At least 10,000 daltons
Greater
Proteins and polysaccharides
Amino acids
Secondary
Epitopes
No
Heart valves, elbow replacements and other
medical appliances
The units of sugars are more limited than the
number of amino acids in proteins
Glycolipids and glycoproteins
Glycolipids
Glycoproteins
Immunogenic
They are eliminated
Plant protein
False
Yes
Enzymatic digestion to create small peptides
that are complexed to MHC molecules to
present to responsive lymphocytes
Yes
27
Immuno PPT Flashcards Unit 1
does it have to undergo antigen processing?
True/False: If an antigen cannot be joined with
an MHC molecule, it would be a poor
immunogen
What is the determinant site?
What cells recognize epitopes?
True/False: Large molecules have few epitopes
Are epitopes sequential in nature?
What is the molecular weight of an
immunogen?
What portion of the immunogen is recognized
in the immune response?
What cells recognize epitopes?
What are the shapes of epitopes?
The folding of 1 chain or multiple chains
bringing certain AAs so they ca be recognize
together is which epitope?
True/False: Epitopes recognized by B cells may
differ from those recognized by T cells
Surface antibodies on B cells react with what
epitopes?
What will trigger B cell activation?
If an immunogen is a protein, B cells will
recognize which structure?
In polysaccharides, do the branch points
contribute to recognition?
Are T cells able to recognize an epitope without
MHC proteins on the surface?
What must an antigen presenting call do to
process an immunogen?
Are T cell epitopes linear or conformational?
If some substances are too small for
recognition, what can they do?
What are haptens?
Can a hapten react with antibody when the
hapten is not complexed to a carrier molecule?
What reaction can occur when haptens crosslink with more than 1 antibody molecule?
What hapten does poison ivy contain?
What does poison ivy cause on the skin?
True/False: Therapeutic drugs and hormones
True
Part of the immunogen that is actually
recognized in the immune response
T or B lymphocytes
False
Yes
At least 10,000 Daltons
Determinate site or epitope
B or T cells
Linear and conformational
Conformational
True
Both linear and conformational
Anything that will cross-link to
immunoglobulin
The primary, secondary, tertiary, or quaternary
structure
Yes
No
It must degrade the immunogen
Linear
Bind to larger molecules to stimulate a response
Nonimmunogenic materials that, when
combined with a carrier, create new antigenic
determinants
Yes, when antibody production is initiated
Precipitation or agglutination reactions
Catechol
Contact dermatitis
True
28
Immuno PPT Flashcards Unit 1
can function as haptens
How are antigens categorized?
What are autoantigens?
Do autoantigens evoke immune responses under
normal circumstances?
What are alloantigens?
What are heteroantigens?
What are heterophile antigens?
What is an adjuvant?
How does an adjuvant function?
What is the only adjuvant approved for clinical
use in the US?
True/False: The hepatitis B vaccine uses an
adjuvant
What antigens determine organ
histocompatibility?
Where are MHC molecules found?
What is the main function of the MHC
molecules?
Why are MHC molecules clinically relevant?
Genes that control expression of molecules are
known as?
True/False: The MHC system is polymorphic
On what chromosome is the coding for the
MHC molecules located?
How many classes of the MHC molecule are
there?
How many locations are class I molecules
coded for?
Where are class II genes situated?
What do class III genes code for?
What are class I and II gene products involved
in?
True/False: Class III proteins are secreted
proteins expressed on cell surfaces
How many copies of chromosome 6 does a
person inherit?
Based on relationship to host
Antigens that belong to the host
No
Antigens from other members of the host’s
species
Antigens from other species, such as other
animals, plants, or microorganisms
Heteroantigens that exist in unrelated plants or
animals that are either identical to or closely
related in structure so that antibody will crossreact with antigen of the other
A substance administered with an immunogen
that increases the immune response
It acts by producing a local inflammatory
response that attracts a large number of immune
system cells to the injection site
Aluminum salts
True
MCH molecules
In all nucleated cells
To bring antigen to the cell surface for
recognition by T cells
They are involved in transfusion reactions, graft
rejection, and autoimmune diseases
Major histocompatibility complex
True
Chromosome 6
3
3
In the D region
Complement proteins and cytokines
Antigen recognition
False
2 copies
29
Immuno PPT Flashcards Unit 1
True/False: MHC genes are codominant
What is a haplotype?
What are in each haplotype?
Polymorphism of MHC has implications in
what?
True/False: Class I MHC molecules are
expressed on all nucleated cells
What cells contain the most class I MHC
molecules?
In what type of organ transplants is HLA testing
not completed?
True/False: HLA-C antigens have the lowest
expression of HLA antigens
What cells have class II MHC molecules?
What are the 3 major class II molecules?
Which class II molecule is expressed at the
highest level?
What are the 3 other class II genes?
Products of the nonclassical class II genes do
what?
What is the main role of class I and class II
MHC molecules?
True/False: T cells can respond to antigens
without being combined with MHC molecules
What molecules do class I molecules present?
True/False: Class I molecules act as watchdogs
for viral antigens
What cells do class II molecules present?
When do class II molecules stimulate CD4
cells?
Where are class I and class II molecules
synthesized?
Where do class I molecules bind peptides?
Peptides that associate with class I molecules
are how many amino acids in length?
What are TAP 1 and TAP 2 proteins responsible
for?
What chain must bind the peptide, MHC-1
peptide complex to transport to the cell surface?
How do class I molecules determine which
antigens an individual will respond?
True
MHC genes that are closely linked and
inherited together as a package
A, B, C, DR, DP, DQ
Organ donation and paternity testing
True
Lymphocytes
Liver transplants
True
B lymphocytes, monocytes, macrophages, and
dendritic cells
DP, DQ, and DR
DR
DM, DN, and DO
Play a regulatory role in antigen processing
Bind peptides within cells and transport them to
the plasma membrane, where T cells can
recognize them in the phenomenon known as
antigen processing
False
Peptides
True
They present antigens to CD 4 cells
In bacterial infections
Rough endoplasmic reticulum
In the rough endoplasmic reticulum
8-10 amino acids
Responsible for the ATP- dependent transport
from the cytoplasm to the lumen of the
endoplasmic reticulum
Alpha chain
Binding affinities
30
Immuno PPT Flashcards Unit 1
Class I molecules complexed to antigens allow
CD 8 positive T cells to continuously do what?
Before class II molecules can bind peptides,
where must they be transported?
Which cells are the most potent activators of T
cells?
What helps to unfold molecules and uncover
functional sites?
What enzyme degrades an invariant chain?
When the invariant chain is degraded, what
fragment is left?
What molecule helps to remove the CLIP
fragment?
Do class I or class II molecules have a closed
end?
Does hydrogen bonding take place along the
length of the peptide in class II molecules?
Can class II proteins accommodate amino acid
side chains?
Once class II protein – peptide complex is
stabilized where is it transported?
What makes up a trimolecular complex?
What results when binding occurs with a T-cell
receptor on a CD4-positive T cell and then T
helper cell recruits and triggers a B-cell
response?
Why is testing for MHC antigens typically
done?
Which test method for MHC antigens is more
accurate?
What is the major role of MHC molecules?
What role do MHC antigens play?
True or False: MHC molecules typically have a
broad binding capacity and small biochemical
differences in these proteins are responsible for
differences seen in the ability to react to a
specific antigen.
True or False: MHC molecules to a particular
vaccine such as hepatitis B do not have the
genetic capacity to respond.
What is an example of the presence of a
particular MHC protein conferring additional
Check cell surfaces for the presence of non-self
antigens
To the endosomal compartment
Dendritic cells
Antigen processing
Protease
Class II invariant chain peptide
HLA-DM
Class I molecules
Yes
Yes
To the cell surface
Class II molecules are responsible for forming
the trimolecular complex that occurs between
antigen, the class II molecule, and an
appropriate T-cell receptor
Antibody formation
Both class I and class II molecules can induce a
response that leads to graft rejection
Molecular method
They determine the types of peptides to which
an individual can mount an immune response
Development of autoimmune disease
True
True
HLA B8 and the increased resistance to HIV
infection
31
Immuno PPT Flashcards Unit 1
protection?
What is made when B cells are stimulated by
antigen?
What are antibodies also known as?
What does “globin” refer to?
Where are immunoglobulins found?
Immunoglobulins are composed of about ____
polypeptide and about ____ carbohydrate.
What are the 5 classes of immunoglobulins?
Immunoglobulins are considered to be the
humoral branch of the ____ _____.
What is the essential role of immunoglobulins?
Are opsonization and complement activation
roles of immunoglobulins?
(T or F) All immunoglobulin molecules share
many common features.
What kind of unit are all immunoglobulin
molecules made up of?
What does the basic four-chain polypeptide unit
consist of?
How are the H chains and L chains held
together?
When subjected to electrophoresis at 8.6,
immunoglobulins appear primarily in the ___
band.
When was the structure of antibodies first
discovered?
What was used to cleave IgG into 3 pieces of
about equal size, each having a sedimentation
coefficient of 3.5 Svedberg units (S) and
representing a molecular weight of
approximately 45,000 to 50,000 Daltons (d)?
What separated the material into two types of
fragments, on which spontaneously crystallized
a 4°C?
What fragment had no antigen-binding ability
and is known to represent the carboxy-terminal
halves of two H chains that are held together by
S-S bonding?
What do antibodies consist of?
What is important in effector functions of
immunoglobulin molecules, which include
opsonization and complement fixation?
Antibodies
Immunoglobulins
Globular shape of protein
Serum portion of blood
85%; 10%
IgG, IgM, IgA, IgD, IgE
Immune response
Antigen recognition and biological activities
related to immune response
Yes
True
Four-chain polypeptide unit
2 heavy (H) chains and 2 light (L) chains
Non-covalent forces and disulfide interchain
bridges
Gamma
1960’s
Proteolytic enzyme papain
Carboxymethyl cellulose ion exchange
chromatography
FC fragment (“fragment crystallizable)
Light and heavy chains that form an FC
fragment, Fab fragment, and a hyervariable
region
FC fragment
32
Immuno PPT Flashcards Unit 1
What are the remaining 2 identical fragments
called?
Because precipitation would not occur if Fab
fragments were allowed to react with antigen,
what was gu100,essed that each fragment
represented?
What does each Fab fragment consist of?
How is each Fab fragment held together?
What was found to cleave IgG at the carboxyterminal side of the interchain disulfide bonds,
yielding one single fragment with molecular
weight of 100,000 d and all the antigen-binding
ability, known as F(ab)2?
What additional fragment was similar to FC
except that it disintegrated into several smaller
pieces?
What is found in the urine of patients with
multiple myeloma?
What type of chains were being secreted by
malignant cells in patients with multiple
myeloma?
Who discovered Bence Jones proteins in 1845?
When heated to 60°C, Bence Jones proteins
____ from urine, but on further heating to 80°C,
they ____.
What were the two main types of L chains in
Bence Jones proteins?
How much amino acid is contained in each L
chain?
What was the region called from positions
number 111 and one where each type had
essentially the same sequence?
What was the amino-terminal end called?
What is the difference between the κ and λ
chains?
(T or F) Both κ and λ L chains are found in all 5
classes of immunoglobulins, but only one type
is present in a given molecule.
What demonstrated the presence of domains
similar to those in the L chains- that is, variable
and constant regions?
What is the first approximately 110 amino acids
at the amino-terminal end?
What are the remaining amino acids typically
divided up into?
Constant regions of the _____ are unique to
Fab fragments (fragment antigen-binding)
One antigen-binding site and that 2 such
fragments were present in an intact antibody
molecule
One L chain and one-half of a H chain
Disulfide bonding
Proteolytic enzyme pepsin
FC’
Bence Jones porteins
L chains
Dr. Henry Bence Jones
precipitated; redissolved
Kappa (κ) and lambda (λ)
Between 200-220 amino acids
Constant region
Variable region
Amino acid substitutions at a few locations
along the chain
True
H chain sequencing
Variable domain
3 or more constant regions with similar
sequences (CH1. CH2, CH3)
H chain
33
Immuno PPT Flashcards Unit 1
each class and give each immunoglobulin type
its name.
What chain does IgG have?
What chain does IgM have?
What chain does IgA have?
What chain does IgD have?
What chain does IgE have?
What is an unique amino acid sequence that is
common to all immunoglobulin molecules of a
given class in a given species?
What are minor variations of amino acid
sequences that are present in some individuals
but not others?
What do the variable portions of each chain that
are unique to a specific antibody molecule
constitute?
What is essential to the formation of the
antigen-binding site?
What is the segment of H chain located between
the CH1 and CH2 regions?
What does the hinge region have a high content
of?
What does the high proline content allow for?
What does the ability to bend in the hinge
region allow for?
What is an example of an effector function?
What chains have a hinge region?
What chains do not have a hinge region?
(T or F) The CH2 domains of the latter two
chains are paired in such a way as to confer
flexibility to the Fab arms.
What do all types of immunoglobulins contain?
Where is the carbohydrate portion located?
What are the functions of the carbohydrate
portion?
(T or F) The basic four-chain structure of all
immunoglobulin molecules does not exist as a
straight “Y” shape.
Is the basic four-chain structure of the Ig folded
into compact globular subunits?
What is the basic four-chain structure of Ig
based on?
What type of bonds stabilize the globular
regions?
γH chain
μ chain
α chain
δ chain
ε chain
Isotype
Allotypes
Idiotype
The aminoterminal ends of both L and H chains
contain these variable regions
Hinge region
Proline and hydrophobic residues
Flexibility
Two antigen-binding sites to operate
independently
Initiation of the complement cascade
Gamma, delta, alpha
Mu and epsilon
True
Four polypeptide chains and a carbohydrate
portion
Between the CH2 domains of the two H chains
Increasing the solubility of Ig, providing
protection against degradation, and enhancing
functional activity of the FC domains.
True
Yes
The formation of balloon-shaped loops at each
of the domains
Intrachain disulfide bonds
34
Immuno PPT Flashcards Unit 1
Within each of the regions or domains, what is
formed when the polypeptide chain is folded
back and forth on itself?
What cylindrical structure is produced when the
folded domains of the H chains line up with
those of the L chains?
What is captured within the barrel by binding to
a small number of amino acids at strategic
locations on each chain (hypervariable regions)?
What is the predominant immunoglobulin in
humans?
What is the percentage of total IgG in humans?
(T or F) IgG has the longest half-life of any Ig
class?
How long is the IgG half-life?
What are the four major subclasses of IgG?
Beta-pleated sheet
Immunoglobulin fold or barrel
Antigen
IgG
75-80%
True
23-25 days
IgG1 (67%), IgG2 (22%), IgG3 (7%), IgG4
(40%)
How do the four major subclasses of IgG differ? In the number and position of the disulfide
bridges between the  chains
Which type of immunoglobulin has the largest
IgG3
hinge region and the largest number of
interchain disulfide bonds, and is the most
efficient at binding complement?
Which type of immunoglobulins have shorter
IgG2 and IgG4
hinge segments and tend to make poor
mediators of complement activation?
What are the major functions of IgG?
Providing immunity for the newborns because
IgG can cross the placenta, fixing complement
coating antigen for enhanced phagocytosis
(opsonization), neutralizing toxins and viruses,
and participating in agglutination and
precipitation reactions
(T or F) All subclasses of IgG appear to be able True
to cross the placenta.
What is the least efficient IgG?
IgG2
What has FC receptors on their surfaces that are Macrophages, monocytes, and neutrophils
specific for the FC region of IgG?
What is the function of FC receptors?
Enhances contact between antigen and
phagocytic cells and increases the efficiency of
phagocytosis
What does IgG have that allows it to enter
High diffusion coefficient
extravascular spaces more readily than other Ig
types?
(T or F) IgG plays a major role in neutralizing
True
toxins and viruses.
IgG is better at ____ reactions than at ____.
Precipitation; agglutination
35
Immuno PPT Flashcards Unit 1
What does precipitation involve that are more
easily brought together by relatively small IgG
molecules?
Small soluble particles
What is defined as the clumping together of
larger particles?
Which Ig is much more efficient at
agglutination than IgG?
What is known as a macroglobulin?
What has a sedimentation rate of 19S, which
represents a molecular weight of approximately
970,000d?
IgM accounts for ____% of all serum.
What is the half-life og IgM?
(T or F) IgM has a shorter half-life than IgG.
What can IgM exist as?
What is the difference between the pentamer
and monomer form?
When a pentamer forms, what holds the five
monomeric units together?
What is a glycoprotein with several cysteine
residues?
What do the cysteine residues serve as?
Agglutination
What is the shape of a IgM configured as a
pentamer?
How many functional binding sites are on a
IgM?
How many functionl binding sites are used on a
IgM?
The ____ valency of IgM antibodies
contravenes the fact that they tend to have ____
affinity for antigen.
Where is IgM mainly found?
Which Ig cannot cross the placenta?
What is the primary response antibody?
What is the first Ig to appear after antigenic
stimulation and the first to appear in the
maturing infant?
(T or F) IgM is synthesized only as long as
antigen remains present.
Are there memory cells for IgM?
Which Ig is known as the secondary response?
What is the primary response characterized by?
IgM
IgM
Macroglobulin
5-10
10 days
True
Monomer or pentamer
Pentamer form is found in secretions; monomer
form occurs on the surface of B cells
J (joining) chain
J chain
Linkage points for disulfide bonds between two
adjacent monomers
Starlike
10
5
High; low
In the intravascular pool and not in other body
fluids or tissues
IgM
IgM
IgM
True
No
IgG
Long lag phase
36
Immuno PPT Flashcards Unit 1
What is the secondary response characterized
by?
What are the functions of IgM?
Shortened lag period and much more rapid
increase in antibody titer
Complement fixation, agglutination,
opsonization, and toxin neutralization
What is the most efficient of all
immunoglobulins at triggering the classical
complement pathway?
(T or F) IgM has multiple binding sites.
The larger number of binding sites makes IgM
more efficient at ____ reactions, especially with
multivalent antigens.
What does IgM form a potent defense against?
Because IgM has a J chain, what can it
occasionally acquire like IgA does?
What does the IgM’s secretory component
allow for?
What does IgM serve as?
What does the presence of membrane IgM
classify lymphocytes as?
What percentage of all circulation
immunoglobulin is IgA?
What is the molecular weight of the H chain?
How much amino acids does the H chain
consist of?
What does IgA appear as?
What is the approximate molecular weight of
IgA?
What are the two subclasses of IgA?
How do IgA1 and IgA2 differ?
How many amino acids are located in the hinge
region and are deleted in IgA2?
What makes IgA more resistant to?
IgM
Where is IgA2 predominately found?
Where is IgA1 mainly found?
IgA2 is found as a ____ along the respiratory,
urogenital, and intestinal mucosa.
What does IgA2 appear in?
Since mucosal surfaces are a major point of
entry for pathogens, what does IgA2 serve?
What does the dimer consist of?
How are the 2 monomers of the dimer held
together?
What is the molecular weight of the dimer?
True
Agglutination
Many bacterial diseases
Secretory component
To traverse epithelial cells and patrol mucous
membranes
Surface receptor for antigen
Mature B cells
10-15%
Between 55,000-60,000
472
Monomer
160,000
IgA1 and IgA2
In content by 22 amino acids
13
Some bacterial proteinases that are able to
cleave IgA1
In secretions at mucosal surfaces
In serum
Dimer
Milk, saliva, tears, and sweat
To keep antigen from penetrating further into
the body
2 monomers
J chain
15,000
37
Immuno PPT Flashcards Unit 1
Where is secretory IgA synthesized?
What form is secretory IgA released in?
What is the molecular weight of a secretory
component?
What is later attached to the FC region around
the hinge portion of the alpha chain?
How many immunoglobulin-like domains, does
the secretory component consist of?
Where is the secretory component derived
from?
What is the molecular weight of the SC
precursor?
Where is the SC precursor found?
What does the SC precursor serve as?
Plasma cells that secrete IgA actually home to
____ ____, where IgA ca bind as soon as it is
released from the plasma cells.
How are epithelial cells traversed?
What occurs after trancytosis?
What can the secretory component act to
facilitate transport of?
What does the secretory component make the
dimer more resistant to?
What is the main function of secretory IgA?
How does secretory IgA act as first line
defense?
What is easily trapped in mucus and then
eliminated by the ciliated epithelial cells of the
respiratory and intestinal tracts?
What does IgA appear not to be capable of?
What may trigger the alternate complement
pathway?
What possess specific receptors for serum and
secretory IgA?
What triggers a respiratory burst and
degranulation of the cell involved?
What can both forms of IgA act as?
What are opsonins?
When was IgD discovered?
In plasma cells found mainly in mucosalassociated lymphoid tissue
Dimeric
About 70,000
Secretory component
Five
Epithelial cells found in close proximity to the
plasma cells
100,000
On the surface of epithelial cells
A specific receptor for IgA
Subeithelial tissue
By transcytosis
A small fragment of SC sttached to IgA is
cleaved to liberate the IgA dimer with the
remaining portion of the SC
IgA to mucosal surfaces
Enzymatic digestion by masking sites that
would be susceptible to protease cleavage
To patrol mucosal surfaces and act as a first line
of defense
Neutralizes toxins produced by microorganisms
and helping to prevent bacterial adherence to
mucosal surfaces
Complexes of IgA and antigen
Fixing complement by the classical pathway
Aggregation of IgA immune complexes
Neutrophils, monocytes, and macrophages
Binding to the sites of IgA
Opsonins
Promoters of phagocytosis
1965
38
Immuno PPT Flashcards Unit 1
IgD was found in a patient with what disease?
What is the percentage of IgD in the total
serum?
What is the molecular weight of IgD?
What is the molecular weight of the H chain?
What appears to have an extended hinge region
consisting of 58 amino acids?
Because of the unusually long hinge region,
what is IgD more susceptible to?
Multiple myeloma
Less than 0.001%
What is the half-life of IgD?
Where are most of the IgD found?
2-3 days
On the surface of immunocompetent but
unstimulated B lymphocytes
IgD
What is the second type of immunoglobulin to
appear?
What may IgD play a role in?
What makes IgD an ideal early responder to
antigen?
What do B lymphocytes bearing only IgM
receptors appear incapable of?
What are those with both IgM and IgD
receptors capable of?
(T or F) IgD may play a role in regulationg Bcell maturation and differentiation.
In the secreted form in the serum, why doesn’t
IgD appear to serve a protection function?
Which immunoglobulin is the least abundant in
the serum?
What is the percentage of IgE in the total
serum?
What is composed of around 550 amino acids
that are distributed over one variable and four
constant domains?
What cells produce IgE and are located
primarily in the lungs and skin?
Does IgE participate in typical immunoglobulin
reactions such as complement fixation,
agglutination, or opsonization?
Is IgE incapable of crossing the placenta?
What happens to IgE shortly after synthesis?
What are the specific surface proteins called?
Where are the high-affinity FC  RI receptors
found?
180,000
62,000
H chain
Proteolysis
B-cell activation
High level of surface expression and its
intrinsic flexibility
IgG response
Responding to T-cell help and switching to
synthesis of IgG, IgA, or IgE
True
It does not bind complement, it does not bind
neutrophils, or macrophages, and it does not
cross the placenta
IgE
0.0005%
The  H chain
Plasma cells
No
Yes
IgE attaches to basophils and tissue mast cells
by means of specific surface proteins
High- affinity FC  RI receptors
Basophils and tissue mast cells
39
Immuno PPT Flashcards Unit 1
Where does the IgE molecule bind at?
Are the antigen-binding sites free to interact
with specific antigen?
Where are mast cells mainly found?
How many receptors are on each mast cell?
What happens when two adjacent IgE
molecules on a mast cell bind specific antigen?
What is induced when vasoactive amines are
released?
What is an allergic reaction called?
What are examples of typical allergic reactions?
What immunoglobulin serves a protective role
by triggering an acute inflammatory reaction
that recruits neutrophils and eosinophils to the
area to help destroy invading antigens that have
penetrated IgA defenses?
What cells play a major role in the destruction
of large antigens, such as parasitic worms, that
cannot be easily phagocytized?
What is the term for one of the first theories to
be formulated in an attempt to explain antibody
diversity in the early 1900s by Paul Ehrlich?
(T or F) Paul Ehrlich postulated that certain
cells had specific surface receptors for antigen
that were present before contact with antigen
occurred.
What happened after an antigen was introduced
and it would select the cell with the proper
receptors?
(T or F) Ehrlich’s side-chain theory laid the
foundation for further hypotheses.
In the 1950s, what idea did Niels Jerne and
Macfarlane Burnet independently support?
What is occurring when individual lymphocytes
are genetically preprogrammed to produce one
type of immunoglobulin and that a specific
finds or selects those particular cells capable of
responding to it, causing them to proliferate?
Where are the surface immunoglobulins IgM
and IgD found?
What would repeated contact with antigen
continually increase?
CH3 domain on the FC region
Yes
In the skin and in the lining of the respiratory
and alimentary tracts
Several hundred thousand
A cascade of cellular events is initiated that
results in degranulation of the mast cells with
release of vasoactive amines (histamine and
heparin)
Type 1 immediate hypersensitivity
Type 1 immediate hypersensitivity
Hay fever, asthma, vomiting and diarrhea,
hives, and life-threatening anaphylactic shock
IgE
Eosinophils
Side-chain theory
True
Combination would take place and then
receptors would break off and enter the
circulation as antibody molecules
True
A clonal selection process for antibody
formation
Clonal selection
On immune-competent but unstimulated B
lymphocytes
A specific lymphocyte pool
40
Immuno PPT Flashcards Unit 1
What explains the kinetics of the immune
response?
Who proposed a solution to the tissue of the
large number of genes required by the clonal
selection theory?
What did Dryer and Bennett suggest?
Clonal selection
There could be a small number coding for the
____ region and a larger number coding for the
____ region.
What happens within lymphocytes?
What controls synthesis of a particular
immunoglobulin, and through a random
selection process these individual segments are
joined to commit that lymphocyte to making
antibody of a single specificity?
(T or F) B cells are genetically preprogrammed
to synthesize very specific antibody.
What antibodies are used for diagnostic testing
and for therapeutic purposes?
Why is the normal response to antigen
heterogeneous?
In 1975, what did Georges Kohler and Cesar
Milstein discover?
In Kohler and Milstein’s technique, what is
fused with an activated B cell?
What is a myeloma cell?
(T or F) Plasma cells normally produce
antibody.
What kind of cell line is chosen for the Kohler
and Milstein’s technique?
What enzyme is deficient in the cell line that is
not capable of producing antibody?
What enzyme is incapable of synthesizing
nucleotides from hypoxanthine and thymidine,
which are needed for DNA synthesis?
What cells are harvested after a mouse is
immunized with a certain antigen?
What cells are combined in the presence of
polyethylene glycol (PEG), a surfactant?
What is produced when the PEG brings about
fusion of plasma cells with myeloma cells?
(T or F) In hybridoma production, only a small
percentage of cells actually fuse and some of
Constant; variable
Dryer and Bennett
The constant and variable portions of
immunoglobulin chains are actually coded by
separate genes
Genetic rearrangements
More than one gene
True
Monoclonal antibodies
A purified antigen has multiple epitopes that
stimulate a variety of B-cell clones
A technique to produce antibody arising from a
single B cell
A myeloma cell
A cancerous plasma cell
True
A particular cell line that is not capable of
producing antibody
Hypoxanthine guanine phosphoribosyl
transferase (HGPRT)
Hypoxanthine guanine phosphoribosyl
transferase (HGPRT)
Spleen cells
Spleen and myeloma cells
Hybridoma
True
41
Immuno PPT Flashcards Unit 1
these are two meloma cells or two spleen cells.
After fusion and cells are placed in culture,
what is contained in the selective medium used?
What is the culture in the HAT medium used to
separate?
What can hybridoma cells synthesize?
Why is one pathway, which builds DNA from
degradation of old nucleic acids, blocked?
The other pathway, which makes DNA from
new nucleotides, is blocked by what?
In hybridoma production, what cells normally
die out?
What cells have the ability (acquired from the
myeloma cell) to reproduce indefinitely in
culture and the ability (acquired from the B cell)
to synthesize nucleotides by the HGPRT and
thymidine kinase pathway?
What antibodies were initially used for in vitro
diagnostic testing and in delivering therapeutic
agents in a variety of diseases?
Define cytokines in two parts.
Cytokines are chemical messengers. several
different cell types produce them.
True or False?
Cytokines have activity modulating effects.
They affect ___________ and
___________ systems. They act through
through activation of cell-bound receptor
proteins.
Induce means__________________.
Cytokines are induced in response to specific
stimuli.
Clarify some example stimuli.
Be Specific.
Cytokines can scale to effect:
Hypoxanthine, aminopterin, and thymidine
(HAT)
Hybridoma cells by allowing them to grow
selectively
Nucleotides in the HAT medium
The myeloma cell line employed is deficient in
the required enzymes HGPRT and thymidine
kinase
Presence of aminopterin in the medium
Myeloma cells and normal B cells
Fused hybridoma cells
Monoclonal antibodies
Cytokines are small soluble proteins that
regulate the immune system’s innate immunity.
They are the adaptive response to infection.
True
Hematopoietic and immune systems
to bring about, produce, or cause
to stimulate the synthesis of (a protein,especiall
y an enzyme) by increasing gene transcription.
bacterial lipopolysaccharides (Lipid A),
flagellin, and other bacterial products.
regulation of growth,
differentiation
gene expression
many different cell types including leukocytes
are effected
42
Immuno PPT Flashcards Unit 1
Define autocrine stimulation.
Define paracrine stimulation.
Do cytokines exert systemic or endocrine
activities always? True or False?
Elaborate the stages of basic cytokine
production by host lymphocytes
Cytokines act individually and separately. True
or False?
What is the result of cytokine expression.
Clarify.
What is a cytokine cascade
What often determines the outcome and the
clinical course of infection?
Massive overproduction plus deregulation in the
immune system creates a cytokine storm. What
terminal effects can be expected?
The cytokines include major families. What are
some major families available for study today.
An Interleukin is a _________ cytokine.
For the cytokines to be classified as
interleukins. What are the three criteria they
must satisfy?
Interleukins are numbered… IL-1 to IL-32
Cytokines were originally thought to act solely
on cells of the immune system. What new
discovery was made about them?
Pleiotropic means…
Can many different cytokines share their
properties?
Many cytokines share receptors. Illustrate some
of the effects or activities.
Name two clinical uses of cytokines.
What conditions could the two variants of
affecting the same cell that secreted it
affecting a target cell in close proximity
False, only occasionally do they do so
the ligation of cell adhesion molecules
the recognition of foreign antigens
False; they do not act alone, but in concert with
many others that are induced during the process
of immune activation.
A network is formed regulating leukocyte
activity.
The cytokine cascade produces a spectrum of
activities that lead to the rapid generation of
innate and adaptive immune responses.
the ability or inability to generate certain
cytokine patterns
shock, multi-organ failure, or even death.
tumor necrosis factors (TNF), interferons (IFN),
chemokines, transforming growth factors
(TGF), and colony-stimulating factors (CSF),
interleukins(IL)
unrelated
They must have had their genes cloned. They
must be inducible in leukocytes. Their
biological activities in inflammatory processes
must be cataloged.
IL-1 to IL-32
They may also act on cells outside the immune
system.
Having many effects ie
The nature of cytokine activity relates to: the
widespread distribution of cytokine receptors
on many cell types, and the ability of cytokines
to alter expression of numerous genes.
Yes, they activate some of the same pathways
and genes.
Some may have overlapping effects.
Some may alter the activity of many of the
same genes.
cytokine antagonists
cytokine receptor antagonists
rheumatoid arthritis, psoriasis, asthma, Crohn's
43
Immuno PPT Flashcards Unit 1
cytokines be usable for?
Will there be future demands placed on the labs
for cytokine assays.
The patterns of cytokine expression can
determine…
A patient has presented defects in the cytokines
or their receptors/signal transduction circuits.
What possible illnesses could be a play in the
patient’s body?
What sort of analyses are recommended for the
patient above?
Inflammation due to the body’s innate immune
response has several criteria. Name four.
The innate immune response is___________
but occurs within hours of first contact with
microorganisms .
What is the main function of the innate immune
response?
Choose several cytokines involved in innate
immune response.
IL-1 has 2 subtypes plus 1 antagonist.
IL-1α facts
IL-1β facts
IL-1 facts
disease, transplantation, and cancer treatments
Defenitely
whether the host will be able to mount an
effective defense against and survive certain
infections.
Numerous immunodeficiency syndromes and
leukemias are theoretically possible.
Genetic and Proteomics.
Assess treatment modalities, effectiveness, and
potential gene replacement therapies
fever, swelling, pain, and cellular infiltrates into
damaged tissues.
nonspecific
To recruit effector cells to area.
interleukin-1, tumor necrosis factor-α,
interleukin-6 chemokines, transforming growth
factor-β, and interferons, both α and β.
IL-1α, IL-1β, and IL-1RA (IL-1 receptor
antagonist)
Proinflammatory
monocytes and macrophages
requires the presence of microbial pathogens,
bacterial lipopolysaccharide's, or other
cytokines.
Intracellular
can be released after cell death
can help attract inflammatory cells to areas
where cells and tissues are being killed or
damaged.
is responsible for most of the systemic activity
fever
activation of phagocytes
production of acute phase proteins
is secreted by monocytes
endogenous pyrogen
induces fever(acute phase response)
acts on the hypothalamus(thermostat)
hypothalamus resets body temp to higher level
bacterial and viral growth is inhibited
44
Immuno PPT Flashcards Unit 1
IL-1 facts part 2
lymphocyte activity is increased
induces the production of vascular celladhesion molecules
chemokines and IL-6 production are increased
Also induces the production of colony
stimulating factors in the bone marrow, thereby
increasing the available number of phagocytic
cells that can respond to the damaged tissues
Define diapedesis
a process,
Chemokines and cell-adhesion molecules attract
and assist leukocytes to enter the inflamed area.
IL-1RA facts
produced by monocytes and macrophages
acts as an antagonist to IL-1
blocks the IL-1 receptor
limits the availability of the receptor for IL-1
turns off the response to IL-1 when no longer
needed
Tumor necrosis factor-α (TNF-α) fact sheet
first isolated from tumor cells
induce lysis in tumor cells
the most prominent member of the TNF
superfamily
at least 19 different peptides that have diverse
biological functions.
both membrane-bound and soluble forms
causes vasodilation
increased vasopermeability
lipopolysaccharide is the main trigger from
gram-negative bacteria
secreted by activated monocytes and
macrophages
activate T cells through its ability to induce
expression of MHC class II molecules
enhances antigen presentation
activates T cells
secreted at high levels, TNF can have
deleterious systemic effects
septic shock can result
large amounts of TNF secreted in response to
gram-negative bacterial infections, causing a
decrease in blood pressure, reduced tissue
perfusion, and disseminated intravascular
Tumor necrosis factor-α (TNF-α) fact sheet II
Tumor necrosis factor-α (TNF-α) fact sheet III
major drawbacks
45
Immuno PPT Flashcards Unit 1
Where can TNF-α be found?
TNF-α has a major role in inflammatory
illnesses. Name 2.
IL-6 fact sheet
Where is IL-6 produced?
Chemokines fact sheet
coagulation.
uncontrolled bleeding
present in rheumatoid synovial fluids and
synovial membranes
Crohn’s disease, with rheumatoid arthritis (RA)
a single protein
triggered by lipopolysaccharide
a pleiotropic cytokine
Affects the inflammation, acute phase reactions,
immunoglobulin synthesis, and the activation
states of B cells and T cells.
Stimulates B cells to proliferate and
differentiate into plasma cells.
Induces CD4+ T cells to produce greater
quantities of both pro- and anti-inflammatory
cytokines.
Chemokines facilitate the extravasation of
leukocytes into the tissues in areas of
inflammation.
facilitate the migration of leukocytes into the
tissues
activate integrins
Allow for the co--localization of multiple cell
types
to the damaged tissue
Help to broaden the response to tissue damage.
Enable the leukocytes to migrate between the
endothelial cells.
The gradient of concentration is important to
leukocytes
that chemokines can make.
abnormal receptors have some protection from
the HIV virus
lymphoid and nonlymphoid cells, including a
variety of normal and transformed cells: T cells,
B cells, monocytes and macrophages,
fibroblasts, hepatocytes, keratinocytes,
astrocytes, vascular endothelial cells, and
various tumor cells.
family of cytokines that enhance motility
Are classified into four families based on the
position of N-terminal cysteine residues.
40 known chemokines
46
Immuno PPT Flashcards Unit 1
Define chemotaxis?
A process that promotes migration of many
types of white blood cells toward the source of
the chemokine.
Chemokines regulate leukocyte activities
include the response to infectious diseases,
autoimmune inflammation, cancer, and the
homing of lymphocytes to all the lymphoid
tissues.
Identify the four chemokine groups and state the alpha, or CXC, chemokines—contains a single
differences.
amino acid between the first and second
cysteines.
beta, or CC, chemokines—has adjacent cysteine
residues.
C chemokines—lacks one of the cysteines.
CX3C—has three amino acids between the
cysteines.
What are chemokines’ key roles in numerous
disease processes?
Initiation, development of inflammatory
processes.
Chemokine drawbacks.
Chemokine receptors are utilized by the HIV
virus to gain access to the cell.
composed of three isoforms: TGF-β1, β2, and
β3
Induced growth arrest in tumor cells in early
studies.
Induces antiproliferative activity
inhibits the activation of macrophages
anti-inflammatory factor for mature T cells.
production by helper T cells
important factor in the establishment of oral
tolerance to bacteria normally found in the
mouth.
autocrine regulator to limit the expansion of
activated cells
cell growth
differentiation
apoptosis,
migration,
the inflammatory response
Transforming growth factor beta (TGF-β) fact
sheet
When active TGF-β regulates….
47
Immuno PPT Flashcards Unit 1
Type I interferons can interfere
with___________. Especially IFN-α and IFN-β.
What cells produce interferons?
Interferons induce….
Type I IFN can activate__________.
Type I IFN enhances ____________.
Type I IFN works against…
Cytokines involved in the _____________are
mainly secreted by T cells, especially T helper
(Th) cells, and affect T- and B-cell function
more directly than was the case with cytokines
in the innate immune response.
helps down-regulate the inflammatory response
when no longer needed
viral replication
dendritic cells
The production of proteins and pathways that
directly interfere with viral replication and cell
division.
natural killer cells
The expression of MHC class I proteins, thus
increasing the recognition and killing of virusinfected cells.
certain malignancies and other inflammatory
processes
adaptive immune response
There are three main subclasses of Th cells.
Identify them.
Th1, Th2, and Treg (T regulatory cells).
What is the function of the T-cell receptor?
Captures antigens and is a target of clonal
expansion of CD4+ T helper cells.
induce high-level expression of IFN-γ.
Th1 cells also secrete IL-2.
IL-2
a. T-cell growth factor
b. drives the growth and differentiation of
both T and B cells
c. and induces lytic activity in NK cells
d. can activate proliferation of Th2 cells
e. helps to generate IgG1- and IgEproducing cells
Th1 cell factsheet
IL12 fact sheet
IFN-γ fact sheet
produced by macrophages and B cells
multiple effects on both T cells and natural
killer (NK) cells
responds to certain stimuli, such as
mycobacteria, intracellular bacteria, and viruses
increases the cytolytic ability of natural killer
(NK) cells.
the principal molecule produced by Th1 cells
It affects the RNA expression levels of more
than 200 genes.
48
Immuno PPT Flashcards Unit 1
Th2 cell factsheet
IL-10 fact sheet
Tregs fact sheet
stimulates antigen presentation by MHC I and
MHC II molecules
regulates theactivation of CD4+ Th1 cells,
CD8+ cytotoxic lymphocytes, NK cells,
bactericidal activities, IL-12R, and IL-18R
primarily responsible for antibody-mediated
immunity
produced by monocytes, macrophages,
CD8+ T cells, and Th2 CD4+ T cells
It inhibits antigen presentation by macrophages
stimulates CD8+ T cells
allergies,
autoimmune diseases,
fighting off parasites
anti-inflammatory and suppressive effects
onTh1
an antagonist to IFN-γ
down-regulator of the immune response.
CD4+ CD25+ T cells
Thymus is their home.
Play a key role in establishing peripheral
tolerance to a wide variety of self-antigens,
allergens, tumor antigens, transplant antigens,
and infectious agents.
Effected by Foxp3 a transcription factor that
causes Treg cells to suppress the activity of
other T cells.
Name the colony stimulating factors(CSF).
colony stimulating factors(CSF) fact sheet
Erythropoietin (EPO) fact sheet
Erythropoietin can improve __________.
Which one is not a symptom of endotoxic
Responsible for inducing IL-10 and TGF-β
expression in adaptive T regulatory 1 (Tr1)
cells in the peripheral circulation.
prevent chronic inflammation.
IL-3
EPO erythropoietin
G-CSF granulocye
M-CSF macrophage
GM-CSF granulocyte
act on bone marrow cells and promote specific
colony formation for the various cell lineages.
regulates RBC production in the bone marrow
primarily produced in the kidneys
oxygenation of the tissues
Dypsia is not a mechanism of Endotoxic Shock.
49
Immuno PPT Flashcards Unit 1
shock?
A. DIC
B. Multi-organ failure
C. Nuetropenia
D. Dypsia
What is the Complement immune system?
Where does the compliment protein
synthesis?
How does the complement system get
activated?
D is the answer
•
Complement is a complex series of
more than 30 soluble and cell-bound
proteins that interact in a very specific
way to enhance host defense
mechanisms against foreign cells.
• It was named by Paul Ehrlich because it
complements the action of antibody in
destroying microorganisms.
• While complement promotes
opsonization and lysis of foreign cells
and immune complexes, chronic
activation can lead to inflammation and
tissue damage. It is the latter that occurs
in autoimmune diseases; therefore,
complement activation needs to be
carefully regulated in order to minimize
tissue damage. In addition to the major
proteins involved in activation, there are
numerous proteins that act as controls or
regulators of the system. The three
pathways for activation of complement
will be discussed along with major
system controls.
• Most plasma complement proteins are
synthesized in the liver, with the
exception of C1 components, which are
mainly produced by intestinal epithelial
cells, and factor D, which is made in
adipose tissue.
• Other cells, such as monocytes and
macrophages, are additional sources of
early complement components,
including C1, C2, C3, and C4.
• Most of these proteins are inactive
precursors, or zymogens, which are
converted to active enzymes in a very
precise order (see Table 6-1).
The complement system can be activated in
three different ways
• The first is the classical pathway, which
involves nine proteins that are triggered
50
Immuno PPT Flashcards Unit 1
•
•
•
•
•
What are the roles of the complement in the
immune defense system?
•
•
•
•
•
What is the immune classic pathway?
•
by antigen–antibody combination.
The second pathway, the alternative
pathway, is an antibody-independent
means of activation of complement.
The third pathway, the lectin pathway, is
another antibody-independent means of
activating complement proteins.
Its major constituent, mannose- (or
mannan-) binding lectin (MBL), adheres
to mannose found mainly in the cell
walls or outer coating of bacteria,
viruses, yeast, and protozoa.
Complement activation seldom involves
only one pathway.
The complement system plays a major
part in the inflammatory response
directed against foreign antigens.
Although the end product of
complement activation is lysis of the
invading cell, many other important
events take place along the way.
Complement fragments act as opsonins,
for which specific receptors are present
on phagocytic cells, thus enhancing the
metabolism and clearance of immune
complexes.
In fact, uptake of immune complexes in
the spleen appears to be complementdependent.
Complement components are also able
to increase vascular permeability, recruit
monocytes and neutrophils to the area of
antigen concentration, and trigger
secretion of immunoregulatory
molecules that amplify the immune
response.
Any deficiencies in the complement
system can result in an increased
susceptibility to infection or in the
accumulation of immune complexes
with possible autoimmune
manifestations.
The classical pathway, or cascade, is the
51
Immuno PPT Flashcards Unit 1
How does the IgG play a role in the classic
pathway?
What are the other substances that can bind
complement directly to initiate the classical
cascade?
How many stages can the complement
activation can be divided and what are they?
main antibody-directed mechanism for
triggering complement activation.
• IgM, IgG1, IgG2, and IgG3 are capable of
activation through the classical pathway.
• IgM is the most efficient, because it has
multiple binding sites; thus, it takes only
one IgM molecule attached to two
adjacent antigenic determinants to initiate
the cascade.
• Two IgG molecules must attach to
antigen within 30 to 40 nm of each other
before complement can bind, and it may
take at least 1,000 IgG molecules to
ensure that two are close enough to
initiate such binding.
• Within the IgG group, IgG3 is the most
effective, followed by IgG1 and then
IgG2.
• Some epitopes, notably the Rh group, are
too far apart on the cell for this to occur,
so they are unable to fix, or activate,
complement.
• In addition to antibody, there are a few
substances that can bind complement
directly to initiate the classical cascade.
• These include C-reactive protein, several
viruses, mycoplasmas, some protozoa, and
certain gram-negative bacteria, such as E.
coli.
• However, most infectious agents can
directly activate only the alternative
pathway.
•
•
•
•
•
Complement activation can be divided
into three main stages, each of which is
dependent on the grouping of certain
reactants as a unit.
The first stage involves C1, which is
known as the recognition unit.
Once C1 is fixed, the next components
activated are C4, C2, and C3, known
collectively as the activation unit.
C5–C9 comprise the membrane attack
complex, and it is this last unit that
completes lysis of the foreign particle.
Figure 6-1 depicts a simplified scheme of
52
Immuno PPT Flashcards Unit 1
the entire pathway.
What is the first recognition unit in the
complement complex?
•
•
•
What is the shape and the function of the
Clq in the complement complex?
•
•
•
•
•
What is the function of the Clq in
recognizing antibodies?
•
What are the Clr and the Cls and how are
they activated?
•
•
•
•
The Recognition Unit: C1qrs
The first complement component to bind
to the cell membrane of the bacterium or
other cell to be destroyed is C1, which
consists of three subunits: Clq, Clr, and
Cls, which are stabilized by calcium.
The complex is made up of one Clq
subunit and two each of the Clr and Cls
subunits.
The (Clr, Cls)2 complex is an S-shaped
structure with several domains of unequal
size.
It is hypothesized that this structure
assumes the shape of a distorted figure
eight, and it wraps itself around the arms
of Clq (see Fig . 6-2).
Clq has a molecular weight of 410,000
and is composed of six strands that form
six globular heads with a collagen-like tail
portion.
This structure has been likened to a
bouquet with six blossoms extending
outward (see Fig. 6-2).
Each of the six “stalks” is composed of
three homologous polypeptide chains—A,
B, and C—that form a triple helix.
Clq “recognizes” the fragment
crystallizable (FC) region of two adjacent
antibody molecules, and at least two of
the globular heads of Clq must be bound
to initiate the classical pathway.
Clr and Cls are both serine protease
proenzymes or zymogens; as binding of
Clq occurs, both are converted into active
enzymes.
Autoactivation of Clr results from a
conformational change that takes place as
Clq is bound.
Mechanical stress transmitted from the
stems as binding occurs opens up the
active site on Clr.
Once activated, Clr cleaves a thioester
53
Immuno PPT Flashcards Unit 1
•
•
•
What is phase 2 of the activation unit and
what does it produce?
•
•
•
•
•
•
•
How is C2 cleaved and form C2a?
•
•
How is C2 converted to C4b and how will they
form the C3 convertase?
•
•
bond on Cls, which activates it.
Activated Clr is extremely specific,
because its only known substrate is Cls.
Cls has a limited specificity, with its only
substrates being C4 and C2.
Once Cls is activated, the recognition
stage ends.
Phase 2, the formation of the activation
unit, results in the production of an
enzyme known as C5 convertase.
Cls cleaves C4 to split off a 77-amino acid
fragment called C4a.
In the process, it opens a thioestercontaining active site on the remaining
part, C4b.
C4b must bind to protein or carbohydrate
within a few seconds, or it will react with
water molecules to form iC4b, which is
rapidly degraded.
Thus, C4b binds mainly to antigen in
clusters that are within a 40 nm radius of
C1.
This represents the first amplification step
in the cascade, because for every one C1
attached, approximately 30 molecules of
C4 are split and attached.
C2 is the next component to be activated.
When combined with C4b in the presence
of magnesium ions, C2 is cleaved by Cls
to form C2a (which has a molecular
weight of 70,000) and C2b (which has a
molecular weight of 34,000) (see Fig. 63A).
This is the only case for the designation
“a” to be given to the cleavage piece with
enzyme activity (see Fig. 6-1).
Binding of C2 to C4b can occur in the
fluid phase, but C4b attached to antigen is
much more efficient in accepting C2. This
serves to keep the reaction localized.
The combination of C4b and C2a is
54
Immuno PPT Flashcards Unit 1
•
•
•
How does C3 important in the activation
pathway and what is it's structure?
•
•
•
•
After the cleavage of C3, what are the
amplification processes?
•
•
•
•
•
•
The membrane attack complex is which phase
known as C3 convertase (see Fig. 6-3B).
This is written as C4b2a to indicate that
the complex is an active enzyme.
The half-life is estimated to be between 15
seconds and 3 minutes, so C3 must be
bound quickly.
If binding does occur, C3 is cleaved into
two parts, C3a and C3b.
C3 serves as the pivotal point for all three
pathways, and cleavage of C3 to C3b
represents the most significant step in the
entire process of complement activation.
C3 consists of two polypeptide chains,
alpha and beta.
The alpha chain contains a highly reactive
thioester group, and when C3a is removed
by cleavage of a single bond in the α
chain, the thioester is exposed.
The remaining piece, C3b, is then capable
of binding to hydroxyl groups on
carbohydrates and proteins in the
immediate vicinity.
The cleavage of C3 represents a second
and major amplification process, because
about 200 molecules are split for every
molecule of C4b2a.
In addition to being required for the
formation of the membrane attack
complex, C3b also serves as a powerful
opsonin.
Macrophages have specific receptors for
C3b.
However, a large number of molecules are
needed for this to occur; hence the need
for amplification.
If C3b is bound within 40 nm of the
C4b2a, then this creates a new enzyme
known as C5 convertase.
Figure 6-3C depicts this last step in the
formation of the activation unit.
Phase 3
55
Immuno PPT Flashcards Unit 1
of the complement cascade?
The cleaving of C5 with deposition of C5b at
another site on the cell membrane constitutes
the beginning of the?
What consists of two polypeptide chains, α and
β, which are linked by disulfide bonds?
C5 convertase consists of what?
C5 convertase splits into what?
C5b attaches to what, forming the beginning of
the MAC?
What is extremely labile, and it is rapidly
inactivated unless binding to C6 occurs?
C6, C7, C8, and C9 do not have what?
Membrane damage is caused by what two
different mechanisms?
What causes a reordering and reorientation of
molecules that results in leaky patches?
When complement proteins are bound,
membrane phospholipids rearrange themselves
into domains surrounding the C5b6789 complex
causing what?
What is able to pass freely out of the cell?
The membrane attack unit begins when?
C6 does what when it binds to C5b?
What attaches to the cell surface?
What binds to C5b6 forming a trimolecular
complex that has a high affinity for lipid
constituents of the cell membrane?
What allows for insertion of the C7 part of the
C5b67 complex into the membrane of the target
cell?
What binds to C7 and exposes a hydrophobic
region that interacts with the cell membrane to
form a small hole in the membrane?
Which protein is inserted into the lipid bilayer?
Lysis can be observed after which protein
binds?
Which protein accelerates the lysis process?
Binding of C8 causes a loss of what from the
cell?
What follows the leakage of potassium from the
cell?
What polymerizes only when bound, and it is
believed that the C5–C8 complex acts as a
catalyst to enhance the rate of reaction?
What forms a hollow, thin-walled cylinder,
membrane attack complex (MAC)
C5
C4b2b3b
C5a and C5b
the cell membrane
C5b
enzymatic activity
channel formation and the binding of
phospholipids
the binding of phospholipids
the integrity of the membrane to be destroyed
Ions
C6 binds to C5b
It stabilizes it
The C5bC6 complex
C7
The binding of C7
C8
C8
C8
C9
potassium
The leakage of amino acids and ribonucleotides
C9
Polymerized C9
56
Immuno PPT Flashcards Unit 1
which constitutes the transmembrane channel?
Pathogens can be destroyed in the absence of
antibody in which pathway?
Which pathway acts as part of innate or natural
immunity?
Which pathway was originally named for the
protein properdin?
What does not initiate this pathway but rather
stabilizes the C3 convertase formed from
activation of other factors?
The alternative pathway does not involve which
proteins?
Which pathway has to get to C3 without going
through C2 and no C4 and without antibodies?
Some Ag’s, like LPS, directly trigger a little of
which protein to be cleaved?
Which protein is unstable, and is the pivotal
molecule to activate?
What protein acts on factor B?
What acts on Bb to create C3Bb?
What binds to host cells to prevent lysis and
inhibits C3 activation?
What •is a FACTOR that becomes a
SUBSTRATE, which acts as an ENZYME on
C3, as part of a complex?
What are the proteins that are unique to the
alternative pathway?
Bacterial cell walls, especially those containing
lipopolysaccharide, fungal cell walls, yeast,
viruses, virally infected cells, tumor cell lines,
and some parasites, especially trypanosomes are
substances that trigger which pathway?
Bacterial cell walls, especially those containing
lipopolysaccharide, fungal cell walls, yeast,
viruses, virally infected cells, tumor cell lines,
and some parasites, especially trypanosomes
can serve as sites for binding what?
What is the first step in the alternative pathway?
In plasma, which protein is not stable?
Water is able to hydrolyze a thioester bond and
thus spontaneously activate a small number of
which molecules?
Once activated, C3 can bind to what in the
alternative pathway?
Binding of C3b to B causes a conformational
change that makes B more susceptible to
the alternative pathway
the alternative pathway
the alternative pathway
Properdin
C2 and C4
the alternative pathway
C3
C3
C3b
Factor D
Factor H
Properdin (Factor P)
Properdin, factor B and factor D
The alternative pathway
C3bBb
The conversion of C3
native C3
C3
factor B
serine proteases
57
Immuno PPT Flashcards Unit 1
cleavage by what?
Bound factor B can be cleaved by which factor?
Factor D is a plasma protein that goes through a
conformational change when it binds to what
factor?
Factor D’s only substrate is bound to which
factor?
What protein cleaves factor B into two pieces:
Ba and Bb?
What complex forms the initial C3 convertase
of the alternative pathway?
C3bBb is then capable of cleaving what
protein?
C3Bb creates an amplification loop that feeds
C3b into which pathway(s)?
The enzyme C3bBb is extremely unstable
unless what binds to the complex?
C3bBb can also cleave what protein, but it is
much more efficient at cleaving C3.?
Which enzyme has a high affinity for C5 and
exhibits C5 convertase activity?
What is the first part of the membrane attack
unit?
Both the alternative and classical pathways are
identical from the cleavage of which protein?
Which pathway provides link between the
innate and acquired immune response?
Which pathway involves nonspecific
recognition of carbohydrates that are common
constituents of microbial cell walls and that are,
importantly, distinct from those found on
human cell surfaces?
What proteins bind to carbohydrates?
What binds to mannose or related sugars in a
calcium-dependent manner to initiate the lectin
pathway?
What are found in glycoproteins or
carbohydrates of a wide variety of
microorganisms, such as bacteria, yeasts,
viruses, and some parasites?
What is a component of many fungi (those
which contain chitin, which is composed of
mannose), protozoa, and some bacteria (those
which have unusual LPS, OPS, or capsules with
mannose)?
What is considered an acute phase protein,
factor D
factor B
factor B
factor D
C3Bb
C3
the classical and alternative pathways
properdin
C5
C3bBb3bP
C5b
C5 to make C5b
lectin pathway
lectin pathway
Lectins
mannose-binding, or mannan-binding, lectin
(MBL)
sugar
Mannose
mannan-binding lectin (MBL)
58
Immuno PPT Flashcards Unit 1
because it is produced in the liver and is
normally present in the serum but increases
during an initial inflammatory response?
Deficiencies of MBL have been associated with
what serious infections?
The structure of what is similar to that of C1q?
MBL is associated with which proteases?
MBL binds to what cellular surface structure?
What is homologous to C1s and causes auto
activity?
What takes the active role in cleaving C4 and
C2?
The functions of which MASPs are unclear at
this time.
Once C4 and C2 are cleaved, the rest of the
lectin pathway is identical to which pathway?
All complement pathways produce which
proteins?
Which proteins are released in a gradient that
increases as it gets closer to the source?
What is also an anaphylaxis protein, constricts
blood vessels to prevent spread?
What cells in the area have receptors (C3aR and
C5aR) on their surface?
What signals the nucleus that they have been
activated?
What happens after the receptors signal the
nucleus that they have been activated?
What is intentional direction movement by
chemonavigation UP the gradient called?
On the way to the bacteria, the phagocyte will
begin to express what?
What causes the neutrophils to stick weakly to
the host epithelium so they can creep along the
surface of the epithelium, seeking its target?
What causes the neutrophils to squeeze through
the blood vessel wall (DIAPEDESIS) to get to
the bacteria?
What ensures that infectious agents and not selfantigens are destroyed and that the reaction
remains localized?
Why are the majority of the control proteins
acts to halt accumulation of C3b?
Which is a glycoprotein that inhibits activation
at the first stages of both the classical and lectin
neonatal pneumonia and sepsis
mannan-binding lectin (MBL)
three MBL-serine proteases (MASPs): MASP1, MASP-2, and MASP-3
MASP-2
MASP-2
MASP-2
MASP-1 and MASP-3
Classical pathway
C3a and C5a
C3a and C5a
C5a
Phagocytes
receptors
Cytoskeletal Reorganization
vector motility
Adhesion molecule receptors
Selectons
Integrins
plasma proteins
Because activation of C3 is the pivotal step in
all pathways
C1 inhibitor, or C1INH
59
Immuno PPT Flashcards Unit 1
pathways?
What glycoprotein’s main role is to inactivate
C1 by binding to the active sites of C1r and
C1s?
What remains bound to the antibody, but all
enzymatic activity ceases when C1 inhibitor, or
C1INH binds?
What inactivates MASP-2 binding to the MBLMASP complex, thus halting the lectin
pathway?
Soluble C4b-binding protein (C4BP) and three
cell-bound receptors, complement receptor type
1 (CR1), membrane cofactor protein (MCP),
and decay accelerating factor (DAF) all inhibit
what?
What is a serine protease that inactivates C3b
and C4b when bound to one of these regulators?
Once bound to CR1, both C4b and C3b can then
be degraded by?
What also acts as a receptor on platelets and red
blood cells and to mediate transport of C3bcoated immune complexes to the liver and
spleen?
What is the most efficient cofactor for factor I–
mediated cleavage of C3b?
How does MCP also help to control the
alternative pathway?
What is capable of dissociating both classical
and alternative pathway C3 convertases?
What portion of DAF is covalently attached to a
glycophospholipid anchor that is inserted into
the outer layer of the membrane lipid bilayer?
What is the reason that DAF can reach C3
convertase sites that are not immediately
adjacent to it?
What is the reason that the presence of DAF on
host cells protects them from bystander lysis
and is one of the main mechanisms used in
discrimination of self from nonself?
What is the principal soluble regulator of the
alternative pathway?
Which factor acts by binding to C3b, thus
preventing the binding of factor B?
C3b in the fluid phase has a hundredfold greater
affinity for which factor than for factor B?
On cell surfaces, C3b preferentially binds which
C1 inhibitor, or C1INH
C1q
C1 inhibitor, or C1INH
formation of C3 convertase in the classical and
lectin pathways
factor I
factor I
CR1
MCP
Since binding of factor B to C3b is inhibited
DAF
The carboxy-terminal
arrangement of DAF in the lipid bilayer allows
DAF mobility within the membrane
because foreign cells do not possess DAF
factor H
factor H
factor H
Factor B
60
Immuno PPT Flashcards Unit 1
factor?
Which factor accelerates the dissociation of the
C3bBb complex on cell surfaces?
Which factor acts as a cofactor that allows
factor I to break down C3b
Which protein is also known as vitronectin?
Which protein interacts with the C5b67
complex as it forms in the fluid phase and
prevents it from binding to cell membranes?
What causes binding of C8 and C9 to still
proceed, but polymerization of C9 does not
occur, and the complex is unable to insert itself
into the cell membrane or to produce lysis?
What also acts to block formation of the
membrane attack complex besides S protein?
What protein’s main function is to bind to C8
and prevent insertion of C9 into host cell
membranes?
What protein plays an important role as part of
the B-cell coreceptor for antigen?
What acts with CD19, and binds complementcoated antigen and cross-links it to membrane
immunoglobulin to activate B cells?
What are more effective at enhancing B-cell
differentiation and production of memory cells
than is antigen by itself?
What cell surface molecule is found on
monocytes, macrophages, neutrophils, and
natural killer cells, specifically binds particles
opsonized with iC3b, a C3b degradation
product?
What receptor plays a key role in mediating
phagocytosis of particles coated with these
complement fragments?
Patients whose white blood cells lack CR3
receptors fail to exhibit functions such as:
What proteins are found on neutrophils,
monocytes, tissue macrophages, activated T
cells, dendritic cells, NK cells, and activated B
cells?
What proteins’ function appears to be similar to
that of CR3, and they may assist neutrophil
adhesion to the endothelium during
inflammation?
What proteins also serve as a means of linking
innate and natural immunity?
Factor H
Factor H
S protein
S protein
S protein
Membrane inhibitor of reactive lysis (MIRL),
or CD59
Membrane inhibitor of reactive lysis (MIRL),
or CD59
CR2
CR2
Immune complexes
CR3 (CD11b/CD18)
CR3
chemotaxis, surface adherence, and aggregation
CR4
CR4
Complement proteins
61
Immuno PPT Flashcards Unit 1
What is a small peptide that causes increased
vascular permeability, contraction of smooth
muscle, and release of histamine from basophils
and mast cells?
C3a, C4a, and C5a are classified as?
What also serves as a chemotaxin for
neutrophils, basophils, eosinophils, mast cells,
monocytes, and dendritic cells
What type of molecules direct neutrophils,
basophils, eosinophils, mast cells, monocytes,
and dendritic cells to the source of antigen
concentration?
Which molecules facilitate phagocytosis and
clearance of foreign substances?
What can be harmful if (1) activated
systemically on a large scale, as in gramnegative septicemia, (2) it is activated by tissue
necrosis such as myocardial infarction, or (3)
lysis of red cells occurs?
Lysis may be another end result of complement
activation, such as in what?
What usually manifests itself in increased
susceptibility to infection and delayed clearance
of immune complexes?
Most of these complement protein conditions
are inherited on an autosomal recessive gene,
and they are quite rare, occurring in what
percent of the general population?
A second deficiency that occurs with some
frequency is that of mannose-binding lectin,
found in what percent of the population?
Lack of what has been associated with
pneumonia, sepsis, and meningococcal disease
in infants?
The most serious deficiency is of what, because
it is the key mediator in all pathways?
Individuals with paroxysmal nocturnal
hemoglobinuria (PNH) have red blood cells that
are deficient in what?
Some studies indicate that a DAF deficiency is
associated with a lack of what?
A deficiency in the what of the DAF molecule
prevents its insertion into the cell membrane?
What prevents insertion of C9 into the cell
membrane by binding to the C5b678 complex,
thus inhibiting formation of transmembrane
anaphylatoxin
anaphylatoxin
C5a
chemotaxin
C4b, C3b, and iC3b
Complement
cold autoimmune hemolytic anemia
Hereditary deficiency of any complement
protein
0.03%
3 – 5%
MBL
C3
DAF
CD59 (MIRL)
glycophospholipid anchor
CD59
62
Immuno PPT Flashcards Unit 1
channels?
Recurrent attacks of angioedema that affect the
extremities, skin, gastrointestinal tract, and
other mucosal surfaces are characteristic of?
What results in excess cleavage of C4 and C2,
keeping the classical pathway going and
creating kinin-related proteins that increase
vascular permeability?
What are the methods most frequently used to
assay individual complement components?
Assays are available for Clq, C4, C3, C5, factor
B, factor H, factor I, C1 inhibitor, and C3a, C4a,
and C5a, but what can they not tell about the
factors?
What is most commonly used to measure lysis,
the end point of complement activation, as a
functional test of complement activity?
What test measures the amount of patient serum
required to lyse 50 percent of a standardized
concentration of antibody-sensitized sheep
erythrocytes?
The CH50 titer is expressed in what kind of
units, which is the reciprocal of the dilution that
is able to lyse 50 percent of the sensitized cells?
What is defined as when the change in lytic
activity per unit change in complement is at a
maximum?
What has been designed as another means of
measuring activation of the classical pathway?
In ELISA assays, what attaches to solid-phase
IgM attached to the walls of microtiter plates?
Antihuman antibody to C9 conjugated to
alkaline phosphatase is the indicator of what?
Alternative pathway activation by means of
what assay?
In the AH50 assay what results from the buffer
system that chelates calcium?
What can detect C3bBbP or C3bP complexes?
(1) Decreased production, (2) increased in vivo
consumption, or (3) in vitro consumption can
lead to what levels of complement components
or activity?
A typical screening test for complement
abnormalities includes testing for which
protein/factor levels?
What occurs after the binding of antigen and
hereditary angioedema
deficiency or lack of C1INH
radial immunodiffusion (RID) and
nephelometry
whether the molecules are functionally active
The hemolytic titration (CH50) assay
The hemolytic titration (CH50) assay
CH50 units
The 50 percent point
ELISA assays
Patient complement
complement activation
AH50 assay
The blocking of classical pathway activation
ELISA assays
Decreased levels of complement components or
activity
C3, C4, and factor B levels
Complement fixation
63
Immuno PPT Flashcards Unit 1
antibody, with uptake of complement, can be
used as an indicator of the presence of either
specific antigen or antibody?
What technique has been used in the detection
of viral, fungal, and rickettsial antibodies?
What test involves a two-stage process: (1) a
test system with antigen and antibody, one of
which is unknown, and (2) an indicator system
consisting of sheep red blood cells coated with
hemolysin, which will cause lysis of the
indicator cells in the presence of complement
If patient antibody is present, it will combine
with the reagent antigen, and what will bind?
If hemolysis is present in the complement
fixation test, this means that no patient antibody
was present, and the test is?
Lack of hemolysis in the complement fixation
test then the test result is?
Which testing results are expressed as the
highest dilution showing no hemolysis?
Why is the use of controls is extremely
important?
What test uses the following controls for
testing: running known positive and negative
sera, an antigen control, a patient serum control,
a cell control, and a complement control?
Complement fixation testing
Complement fixation testing
complement
negative
positive
Complement fixation testing
to ensure the accuracy of test results
Complement fixation testing
64