Shared Flashcard Set

Details

Immunology & Microbiology
Fall 2009
412
Other
Professional
12/12/2009

Additional Other Flashcards

 


 

Cards

Term
IgE is induced by allergen and bound via its Fc receptor to mast cells and eosinophils.  After encountering the antigen again, the fixed IgE induces degranulation and release of mediators (e.g. histamine)
Definition
Type I Hypersensitivity
Term
Antigens on a cell surface combine with antibody, which leads to complement-mediated lysis or other cytotoxic membrane damage
Definition
Type II Hypersensitivity
Term
Ag-Ab immune complexes are deposited in tissues, complement is activated, and PMN cells are attracted to the site, causing tissue damage
Definition
Type III Hypersensitivity
Term
T cells, sensitized by an antigen, release cytokines upon second contact with same antigen.  The cytokines induce inflammation and activate macrophages
Definition
Type IV Hypersensitivity
Term

  1. Vascular Adherence (cell rolling)
  2. Diapedesis - process of neutrophils moving through EC into ECM
  3. Activation - increased granules
  4. Phagocytosis and Killing

Definition
Phagocyte (neutrophil) Response to Infection
Term

Characteristic of neutrophils

 

Contain

Cationic proteins

Defensins (play a role in killing)

myeloperoxidase

Definition
Characteristics of Primary Neutrophil Granules
Term

Specific for Mature Neutrophils

 

Contain

lysozyme

NADPH

Lactoferrin (reduce ability to use Fe)

B-12 binding protein (reduce ability to use B-12)

Definition
Characteristics of Secondary Granules
Term

Receptor Mediated

 

  1. Attachment
  2. Pseudopod Extension
  3. Phagosome Formation
  4. Granule Fusion
  5. Phagolysosome Formation

Definition
Steps for Phagocytosis
Term
Damage to Microbial membranes
Definition
Function of Cationic Proteins
Term
Hydrolyses mucopeptides in the cell wall
Definition
Function of Lysozyme
Term
Deprives pathogens of iron
Definition
Function of Lactoferrin
Term
Digest killed organisms
Definition
Function of Hydrolytic enzymes (proteases)
Term

Fragments of complement proteins released during activation.

 

Results in increased vascular permeability and attracts leukocytes.

Definition
Anaphylatoxins
Term
Bacterial toxins released from damaged cells.
Definition
Endotoxins
Term

CD8

 

T cells that kill other cells

Definition
Cytotoxic T Cells
Term

Lysis of bacteria or cells by insertion of membrane attack complex derived from compliment activation

 

Definition
Cytolysis
Term
A process whereby phagocytic cells are attracted to the vicinity of invading pathogens
Definition
Chemotaxis
Term

Immunity in which the participation of lymphocytes and macrophages is predominant.

 

Generally applied to Type IV Hypersensitivity

Definition
Cell-mediated (cellular) immunity
Term
B cells are the precursors of plasma cells that produce antibody
Definition
B Cellsl
Term
A substance that can react with an antibody
Definition
Antigen (Ag)
Term

A protein produced as a result of interaction with antigen.

 

The protein has the ability to combine with the antigen that stimulated its production.

Definition
Antibody (Ab)
Term
Variants of a single genetic locus
Definition
Alleles
Term
Molecules that mediate the binding of cells to other cells or ECM (such as fibronectin)
Definition
ADhesion Molecules
Term
Large granular lymphoid cells with no known antigen-specific receptors.  The are able to recognize and kill certain abnormal cells and also activate the innate response
Definition
NK cells
Term
A phagocytic blood cell that matures into a tissue macrophage
Definition
Monocyte
Term

A phagocytic mononuclear cell found in tissues and at site of inflamation.

 

Also an accessory role is an antigen-presenting cell (APC)

Definition
Macrophage
Term
Low MW proteins that stimulate leukocyte movement
Definition
Chemokines
Term

Characterized by a multilobed nucleus

 

Phagocytic for bacteria and other particles

Definition
Polymorphonuclear Cell (PMN)
Term
Cluster of gen es in close proximity that encodes the histocompatibility antigens
Definition
Major Histocompatability Complex
Term
A molecule that is not immunogenic by itself but can react with a specific antibody
Definition
Hapton
Term
A set of plasma proteins that is the primary mediator of antigen-antibody reactions
Definition
Complement
Term
Induction of immunity by injecting a dead or attenuated form of a pathogen
Definition
Vaccination
Term
A thymus-derived cell that participates in a variety of cell-mediated immune reactions
Definition
T cell
Term
A terminally differentiated B cell that secretes antibody
Definition
Plasma Cell
Term
The coating of an antigen or particle by sustances (such as Ab, complement, fibronectin, etc.) that facilitate phagocytosis
Definition
Opsonization
Term

A substance capable of enhancing phagocytosis.

 

Antibodies and complement are the two main opsonins.

Definition
Opsonin
Term
The end product of activation of the complement cascade, which makes holes in the membrane of cells, resulting in lysis.
Definition
Membrane Attack Complex
Term

Monomuclear cell containing a densely packed nucleus and a small rim of cytoplasm.

 

Includes T cells and B cells.

Definition
Lymphocyte
Term

Low MW proteins elaborated by infected cells.

 

Protect noninfected cells from viral infection.

 

Cytokines that also have immunomodulating functions.

Definition
Interferon
Term
Local accumulation of fluid and cells after injury or infection
Definition
Inflammation
Term

Subdivision based on structural differences

 

IgG

IgA

IgM

IgE

IgD

Definition
Immunoglobulin Class
Term
A subdivision of the classes of immunoglobulins based on structural differences.
Definition
Immunoglobulin Subclass
Term
A glycoprotein that functions as antibody.
Definition
Immunoglobulin
Term
Protection azquired by deliberate introduction of an antigen into a responsive host.
Definition
Adaptive Immunity
Term
Nonspecific resistance not acquired through contact with antigen
Definition
Innate Immunity
Term

Development of resistance to a foreign substance

 

Can be antibody-mediated, cell-mediated, or both

Definition
Immune Response
Term
Pertaining to Immunity in a body fluid and used to denote immunity mediated by antibody and complement
Definition
Humoral Immunity
Term
Sharing transplantation antigens
Definition
Histocompatible
Term
Site on an antigen recognized by an antibody
Definition
Epitope
Term
Developing T cells found in thymus
Definition
Thymocytes
Term

  1. To produce, maintain, and distribute lymphocytes
  2. To act as a filter
  3. To return excess water from the interstitial space back to the general circulation

Definition
Functions of Lymphoid System
Term

  1. Start as pockets rather than tubes
  2. Larger diameter
  3. Have thinner walls
  4. Flat and irregular in section

Definition
How do lymphatic capillaries differ from blood capillaries?
Term

Special lymphatic capillaries in small intestine

 

Transport lipids from digestive tract

Definition
What are lacteals
Term

from the right lymphatic duct empties into the right subclavian vein

 

from the thoracic duct empties into the left subclavian vein

Definition
Where do all lympatics empty into?
Term
CD66
Definition
CD Markers of Neutrophils
Term
CD 14
Definition
CD Markers of Macrophages
Term

Presence of CD 56 and CD 16

 

Absence of CD 3

Definition
CD Markers of NK cells
Term

CD 8

 

binds to MHC class I molecules

Definition
CD Markers of Cytotoxic T Cells
Term

CD 4

 

binds to MHC class II molecules

Definition
CD Markers on Helper T Cells
Term

Distributed on all nucleated somatic cells

 

Present peptide antigens to CD 8 T cells

Definition
MHC class I
Term

Distributed on antigen-presenting cells and activated T cells

 

Present peptide antigens to CD 4 T cells

Definition
MHC class II
Term

Classical Pathway

 

Lectin Pathway

 

Alternative Pathway

Definition
3 Pathways of Complement Activation
Term

IgM and IgG attach to pathogen

 

C1 binds to Ab

 

C3 is cleaved by C3 Convertase into C3a and C3b

Definition
Describe the Classical Pathway of Complement Activation
Term

Mannose-binding lectin binds to sugar residues found in microbial surface polysaccharides

 

Then the pathway follows the classical pathway from the cleaving of C3

Definition

Describe the Lectin Pathway of Complement Activation

Term
Many different substances activate C3 cleavage
Definition

Describe the Alternative Pathway of Complement Activation

Term

Recruitment of Inflammatory Cells

 

Opsonization

 

Membrane Attack Complex

Definition
3 Possibilities of cell Death from Complement
Term

  1. Neutrophil activation
  2. neutrophil adhesion
  3. promotes diapedesis and chemotaxis
  4. monocyte activation to produce IL-1 and IL-6
  5. Mast Cell degranulation

Definition
Biological Effects of C5a
Term
30+
Definition
How many complement proteins are there?
Term

C3 is cleaved by C3 convertase into C3a and C3b

 

C3a is an anaphylatoxin

 

C3b leads to the MAC or Opsonization

Definition
Why is C3 important in the complement cascade?
Term

Predisposition to Systemic Lupus Erethymetosis

 

Opsonization of immune complexes help keep them soluble, deficiency results in increased precipitation in tissues and inflammation.

Definition
What is the disease and mechanism associated with a deficiency of C1, C2, and C4?
Term

Susceptibility to bacterial infections.

 

Lack of opsonization and inability to utilize MAC

Definition

What is the disease and mechanism associated with a deficiency of C3?

Term

Susceptibility to Gram Negative infections

 

Inability to attack outer membrane

 

 

Clinical manifestation is increased temperature and respiratory rate and decreased BP

Definition

What is the disease and mechanism associated with a deficiency of C5-C9?

Term

Two chains (a and b) that have a transmembrane portion and a distal CDR.

 

This attaches to the MHC markers on a cell surface.

Definition
Describe the structure and function of T Cell receptors
Term

  1. The native protein is denatured
  2. Peptides are cut and placed in MHC "cradle"
  3. MHC is exocytosed to the plasma membrane
MHC class I has b-2 microglobulin (non-covalently linked) and presents to CD8 T cells

MHC class II presents to CD4 T cells
TH1 activates macrophages
TH2 activates B cells

Definition
Describe the structure and process of antigen presentation with MHC markers
Term
Human Leukocyte Antigen (HLA)
Definition
What are MHC markers also known as?
Term

  • Progenetor cells give rise to many lymphocytes of different specificity
  • An antigen comes in contact with a lymphocyte of correct specificity
  • Clonal expansion due to being activated by presentation with antigen

Definition
Describe the process of clonal selection and differentiation.
Term

Complementary Determinant Region

 

The domain on an immunoglobulin or T cell receptor that is variable and includes the active site.

Definition
What is the CDR portion of immunoglobulins and T cell receptors?
Term

Ab sees and binds to native conformation

 

T cell recognition requires processing and is going to be presented on an MHC molecule

Definition
What is different about the peptide when it is presented by surface immunoglobulins as opposed to MHCs?
Term

Type 1 Helper T cell

 

INF-g released by innate system cells

 

activates macrophages or cause B cells to switch to IgG

 

Promotes bacterial clearance

Definition
TH1
Term

Type 2 Helper T cells

 

IL-4 is released

 

Activate mast cells and eosinophils and cause B cells to switch production to IgE

 

Aids in response to parasites

Definition
TH2
Term

pentamer

 

primary immune response

Definition
Characteristics of IgM
Term

dimer

 

prevent attachment of pathogens to EC

Definition

Characteristics of IgA

Term

monomer

 

primary and secondary immune response

Definition

Characteristics of IgG

Term

monomer

 

binds to mast cells and basophils to cause them to release histamine

Definition

Characteristics of IgE

Term

monomer

 

attached to surface of B cell

 

important in B cell activation

Definition

Characteristics of IgD

Term

Somatic recombination

 

Point mutations

 

Class switching

Definition
Mechanisms of Antibody Diversity
Term

Neutralization

 

Agglutination

 

Precipitation

 

Complement Fixation

Definition
Mechanisms of antibody action
Term

[image]Variable region on the N-termini

Fc region is constant and may have transmembrane regions on C-termini if on B cell

 

Heavy Chains are 50 kD each

Light Chains are 25 kD each

Definition
Describe the structure and function of antibodies/immunoglobulin
Term

Source: Macrophages (IFN-a), fibroblasts (IFN-b)

 

Function: Antiviral

Definition
IFN-a, -b
Term

Source: T Cells and NK Cells

 

Function: Activateion of macrophage and TH1 differentiation

Definition
IFN-g (interferon)
Term

Source:  Macrophages and T cells

 

Function:  Cell activation, fever, cachexia, antitumor

Definition
TNF-a
Term

Source: T cells

 

Function:  Activates PMNs

Definition

TNF-b

Term

Source:  Macrophages

 

Function:  Cell activation, fever

Definition
IL-1
Term

Source:  T cells

 

Function:  T cell growth and activation

Definition
IL-2
Term

Source: T cells and mast cells

 

Function: B cell proliferation and switching to IgE

TH2 differentiation

Definition
IL-4
Term

Source: Macrophages

 

Function: Differentiation of T cells

activation of NK cells

Definition
IL-12
Term
Natural immune response
Definition
This includes immunological barriers and the innate immune response
Term
Active and Passive
Definition
These are the two main buckets of the acquired immune response
Term
Passive
Definition
Host is not actively involved in, you just acquire due to transfer of immunoglobulins
Term
Natural and Artificial
Definition
2 Parts of Passive Immunity
Term
Passive Artificial
Definition
Giving someone antibodies against rabies is an example of what immunity?
Term
Passive Natural
Definition
Passing immunity from the mother to fetus or through breastfeeding is an example of which immunity?
Term
Active Artificial
Definition
Receiving a flu shot would be an example of which immunity?
Term
Natural Active
Definition
When exposed to an antigen for a second time and memory B and T cells are activated against it, this type of immunity is?
Term
Vascular

Cellular

Humoral
Definition
What are the three main levels of acute inflammatory response?
Term
IgA
Definition
Which antibody is naturally acquired through colostrum from mother?
Term
Immediate Protection
Definition
What is an advantage of passive immunity?
Term
No long term protection

Serum sickness

Type III Hypersensitivity

Transmissino of Hepatitis and AIDS

GVHD
Definition
What are the disadvantages of passive immunity?
Term
Attenuated organisms

Killed Organisms

Subcellular Fragments

Toxins
Definition
What are 4 methods in which artificial active immunity can be made from?
Term
When T cells are removed from the thymus and B cells are removed from the bone marrow, then they are inactivated to produce tolerance.
Definition
What is central tolerance?
Term
Lack of response due to the absence of signals
Definition
What is peripheral tolerance?
Term
Autoimmunity
Definition
What is the loss of tolerance?
Term
Molecular Mimicry
Definition
A mechanism by which we see autoimmune diseases develop. Molecules that look like self. Therefore host cells are attcked by immune system.
Term
Rheumatic fever
Definition
This is an autoimmune disease that follow strep throat.
Term
Severe Combined Immune Deficiency
Definition
What does SCID stand for?
Term
The thymus is not developed properly

It is a T Cell disease
Definition
What is DiGeorge Disease
Term
Lack of IgG development

Lack of B Cell development
Definition
What is Brutons Disease?
Term
The inability of cells to be phagocytic
Definition
What is Chediak-Higashi Disease?
Term
Pregnancy

Blood Transfusion

Previous Transplant
Definition
What are the three ways that patients can be sensitized to HLA?
Term
Hyperacute Rejection
Definition
This is where the body already has antibodies against transplant.
Term
Acute Rejection
Definition
This is the rejection where HLA sensitization produces HLA specific antibodies and T cells are generated.
Term
Graft versus Host Disease
Definition
What does GVHD stand for?
Term
False

It does increase
Definition
T/F: There is no evidence that the frequency of cancer rises with patients with immunodeficiencies.
Term
Tumor associated Antigens
Definition
These antigens are similar to viral infection and create defective signaling that will not activate T cells properly.
Term
Edward Jenner
Definition
Who developed the smallpox vaccination?
Term
Group A Strep
Definition
What does GAS stand for?
Term
Sniffing of small pox crust
Definition
In 3000 BC Egyptians and 2000 BC in China, sniffed this to be immunized.
Term
Variolation
Definition
The Turks introduced this in 1500 BC
Term
Rabies
Definition
This vaccine was introduced in 1885 by Pasteur
Term
Diphtheria and Tetanus
Definition
This vaccine was introduced in the 1920's
Term
Pertussis
Definition
Vaccine for this was introduced in 1934
Term
MMR by Sabin Polio
Definition
What vaccine was developed in the 1960's and by whom?
Term
Smallpox
Definition
This disease is caused by the variola virus
Term
Activate immune response immediately

Produce antibodies quickly

Prevent disease from occurring because the antigen is eliminated rapidly
Definition
A memory B cell or T cell circulating through the body will elicit which three responses when recognizing antigen?
Term
Live attenuated virus
Definition
This vaccine is made from a virus or bacteria that has to replicate to work.
Term
Inactivated immunization
Definition
Made from a virus or bacteria, can fractionate, and organisms do not replicate.
Term
True
Definition
T/F: A toxoid takes a toxin and chemically alters it so it loses its activity but can still illicit an immune response
Term
Polysaccharide
Definition
This vaccine type stimulates T cell independent immunity. It stimulates B cells without T cells, is short lived and needs boosters.
Term
Conjugate polysaccharide
Definition
This vaccine type stimulates T cell dependent immunity. T helper cells involved, produces memory and results in booster effect upon subsequent exposure.
Term
Guillain-Barre Syndrome (GBS)
Definition
What is a known complication of the H1N1 vaccine?
Term
True
Definition
T/F: Gullain-Barre Syndrome is a neurological disease
Term
True
Definition
T/F: Humoral factors and cell-mediated immune phenomena have been implicated in the damage of myelin and or myelin-producing Schwann cells.
Term
Vaccinations

Epidural anesthesia

Thrombolytic agents
Definition
List three events that GBS follows
Term
Campylobacter
Definition
What is the infection that is known to hae the highest prevalence of GBS if infected with it before GBS?
Term
Immunosupressent Drugs
Definition
What is the primary method of treatment for GBS?
Term
Monoclonal response
Definition
A single cell producing a single antibody specificity
Term
Cleaves both Fab regions and the Fc region
Definition
How does papain cleave IgG?
Term
Yes they can. However, effectiveness is quite limited.
Definition
When papain cleaves an IgG molecule, can Fab and the Fc regions still bind?
Term
Pepsin digestion
Definition
This separates the Fc fragment from the Fab regions and leaves the Fab regions bivalent. Therefore it cannot bind complement anymore.
Term
True
Definition
T/F: A papain digested IgG could neutralize a toxin.
Term
True
Definition
T/F: Could a pepsin digested IgG neutralize?
Term
False

The macrophage would not be able to recognize
Definition
T/F: A pepsin or papaindigested fragment can opsonize?
Term
Myeloma cells
Definition
What is the spleen cells in a laboratory fused with to make an immortal cell line?
Term
Hypervariable regions directing involved with antigen binding
Definition
Humanized Monoclonal Antibodies

Which part of the mouse antibody is not replace with humanized counterparts to create humanized monoclonal antibodies?
Term
True
Definition
T/F: In a chimeric antibody, the variable light and variable heavy chains are xenographic in nature.
Term
Grafted CDR
Definition
What type of humanized monoclonal antibody has a human framework, but xenographic CDR?
Term
Monoclonal antibodies
Definition
Hybridomas and recombinant DNA technologies are two ways of producing ____ _____.
Term
Bacteriophage
Definition
When producing monoclonal antibodies through recombinant technology, what vector is usually used to accomplish this?
Term
False

Pepsin digestion
Definition
Toxins can be coupled to antibodies through papain digestion.
Term
False

OKT3 is against CD3 T cells
Definition
T/F: OKT3 is a mouse monoclonal antibody that is against CD4 T cells.
Term
OKT3
Definition
What monoclonal antibody would one receive if they were rejecting a transplant?
Term
Shuts down ALL the T cells
Definition
Why does OKT3 cause patients to be severely immunosuppressed?
Term
Breast
Definition
Herceptin is a humanized antibody used to treat what type of cancer?
Term
B-cell leukemia

(Chronic lymphocytic leukemia)
Definition
Campath is a humanized monoclonal antibody used against what?
Term
True
Definition
T/F: Humira is a monoclonal antibody used to treat rheumatoid arthritis by acting against cytokine TNF
Term
True
Definition
T/F: Enbrel is a monoclonal antibody used to treat Rheumatoid arthritis by blocking TNF.
Term
T cells
Definition
In transplant rejection, the organ transplanted into the recipient is attacked by what cell in the immune system?
Term
When bone marrow is transplanted, the T cells in the transplant attack the recipient's tissues.
Definition
What is graft versus host disease (GVHD)?
Term
HUman Leukocyte Antigens
Definition
What does HLA stand for?
Term
HLA-A

HLA-B

HLA-C
Definition
What are three HLA class 1 proteins that are important in transplantations?
Term
HLA-DR

HLA-DQ

HLA-DP
Definition
What are the three HLA class II proteins that are important in transplants?
Term
B lymphocytes

Monocytes

Dendritic Cells
Definition
HLA class II molecules are found on what cells?
Term
True
Definition
T/F: HLA class I molecules have an alpha transmembrane chain and a beta-2-microglobulin.
Term
True
Definition
T/F: HLA class II molecules have an alpha and a beta transmembrane chain.
Term
B. Light
Definition
Kappa and Lambda refer to _____ chains of immunoglobulins?

A. Heavy
B. Light
C. Disulfide Bond
D. None of the above
Term
6
Definition
Which chromosome encodes for the HLA genes?
Term
HLA-A
Definition
Which HLA gene is encoded most distal from the centromere?
Term
The tope of the head and the antlers
Definition
Which part of the "Bullwinkle model" contain(s) polymorphisms that govern peptide binding?
Term
Previous rejected organ allografts

Previous pregnancy

Previous blood transfusion (WBC)
Definition
How could preformed anti-HLA antibodies be present in renal transplant candidates?
Term
Hyperacute rejection; graft thrombosis; graft loss

Acute humoral rejection, delayed graft function, early graft loss
Definition
Why are donor reactive HLA specific IgG antibodies a high risk to transplants?
Term
lymphocyte crossmatch

antibody screening tests
Definition
What are two tests that are run to assess HLA sensitization in a patient?
Term
It is a test to ensure that the transplant recipient does not have antibodies.
Definition
In a lymphocyte crossmatch, what is being done?
Term
amos modified NIH XM
cell mediated lysis
Definition
In this type of lymphocyte crossmatch, anti HLA-A, B, or C antibodies are detected, and it causes the cell to lyse.
Term
anti-human globulin (AHG) augmented XM
complement mediated lysis
Definition
What type of lymphocyte crossmatch would detect low levels of IgG anti-HLA or non-complement binding antibodies?
Term
Flow cytometric T and B cell XM

Complement Independent

Able to detect HLA II antibodies
Definition
What type of lymphocyte crossmatch is similar to AHG but is even more sensitive and is not complement-dependent and uses fluorescence?
Term
Least: Amos-Modified NIH XM

Anti-Human Globulin augmented XM

Most: Flow Cytometric T&B cell XM
Definition
Rank the lymphocyte crossmatch from least sensitive to most sensitive?
Term
Dendritic cells from donor migrate to the spleen where they activate effector T cells.

Effector T cells migrate to graft via blood.

Graft destroyed by effector T cells.
Definition
What happens if a kidney used for transplantation is recognized as foreign to the recipients immune system?
Term
48 hours

Yes, HLA matching benefit

Yes, HLA matching is used in allocation
Definition
What is the kidney's max cold time and HLA matching benefit and is HLA matching used in allocation?
Term
24 hours

No HLA matcing benefit

No, HLA matching in allocation
Definition
What is the pancreas's max cold time and HLA matching benefit and is HLA matching used in allocation?
Term
24 hours

No HLA matching benefit

No HLA matching in allocation
Definition
What is the liver's max cold time and HLA matching benefit and is HLA matching used in allocation?
Term
4-5 hours

Yes, HLA matching benefit

No, HLA matching in allocation because it is so rare.
Definition
What is the heart max cold time and HLA matching benefit and is HLA matching used in allocation?
Term
6-8 Hours

Yes, HLA matching benefit

No HLA matching in allocation
Definition
What is the lung's max cold time and HLA matching benefit and is HLA matching used in allocation?
Term
Indefinite

Yes, HLA matching benefit

Yes HLA matching in allocation
Definition
What is the blood stem cell's max cold time and HLA matching benefit and is HLA matching used in allocation?
Term
Autologous Transplantation

Breast Cancer uses this
Definition
What is the graft called when it is within the same individual or autotransplantation?
Term
Syngeneic Graft

Most ideal
Definition
What is the graft called that uses identical twins (isotransplantation)?
Term
Allogeneic Graft

Most common, what Dr. Fowler was a donor for
Definition
What type of graft is non-identical, allotransplantation?
Term
Xenogeneic graft
Definition
What type of graft occurs between species, xenotransplantation?
Term
Cytotoxic Agents

Agents interfering with gene expression

Agents interfering with intracellular signaling

Agents interfering with intercellular signaling
Definition
What do immunosuppressive agents do in the body?

4 things
Term
False

The recipient's T cells attack... leads to graft rejection
Definition
T/F: In kidney tranplantation, the donor's T cells attack the transplant.
Term
True

Leads to GVHD
Definition
T/F: In bone marrow transplantation, the T cells in the transplant or graft attack the recipient's tissue.
Term
Conditioning regimen
Definition
A regimen in which the transplant recipient is prepared for bone marrow transplantation is called _____.
Term
The antigen in the HLA of the recipient is the one recognized by the T cells of the donor.
Definition
What is the antigen being presented in GVHD?
Term
By T cell recognitionof host alloantigens.

Term alloantigens means same thing as HLA MHC.
Definition
How is GVHD initiated?
Term
NK cells, CD8+ T cels, cytokines
Definition
Effector cells are cells from the donor. What are a few that are prevalent in the recipient post engraftment?
Term
Epithelial cell necrosis, skin, liver, gastrointestinal tract

Characterized by skin rash, jaundice, and diarrhea
Definition
What are some sign of acute GVHD?
Term
Minor histocampatibility antigens are governing the immune response
Definition
What is the term when all HLA are matched and we still see GVHD?
Term
Adv: Donor availability and no GVHD

Dis: potential infusion of malignant cells
Definition
What is an advantage and disadvantage of autologous transplant?
Term
Adv: No malignant cells infusion, decreased rate of GVHD

Dis: Donor availability and limited GVL
Definition
What is an advantage and disadvantage of syngeneic transplant?
Term
Adv: No malignant cells in infusion, GVL

Dis: potential GVHD, potential graft rejection, donor availability, limited by age and co-morbidities
Definition
What is an advantage and disadvantage of allogeneic transplant?
Term
The goal and the rationale is to attempt to eradicate all tumor cells prior to bmt and use hemopoetic cells to rescue the patient
Definition
What does it mean to maximize conditioning?
Term
The goal and the rationale is to use donor stem cells for tolerance and donor T cells and NK cells for GLV/GVTumor or otherwise abnormal host cells
Definition
What does it mean to minimize the conditioning?
Term
False

They would most likely undergo maximal conditioning
Definition
T/F: A patient who is young and healthy would most likely undergo minimal conditioning.
Term
No GVHD is noticed
Definition
Is there a sign of GVHD in stage 0
Term
Stage IV
Definition
What is the worst stage of GVHD?
Term
allele mismatch
single class I mismatch and single class II
mismatch
multiple class i mismatch
class i and class ii mismatch
Definition
rank the probability of survival from highest to
lowest of the following:
multiple class i mismatch
class i and class ii mismatch
single class i mismatch
single class ii mismatch
allele mismatch
Term
class i and class ii mismatch
single class ii mismatch
multiple class i mismatch
single class i mismatch
allele match
Definition
rank the propability of gvhd based on mismatching
from highest to lowest:
allele match
multiple class i mismatch
class i and class ii mismatch
single class i mismatch
single class ii mismatch
Term
inhibits dna synthesis....interfers with and inhibits tcell profileration
Definition
what is azathioprene?
Term
interfering with dena replication and rna
transcription
Definition
what is cyclophosphamide?
Term
dihydrofolate reductase inhibitor..inhibits dna
synthesis by inhibiting the production of thymidine
Definition
what is methotrexate?
Term
antibody against tcells.....depleting tcells inducing
complement lysis and uptake...modulates and
masks cell surface receptors for t&b lymphocytes...
Definition
what is thymoglobulin?
Term
interfers with signal tranduction from tcr to inhibit t
cell activation and reduces transcription of cytokine
genes
Definition
cysclosporine?
Term
block independent signaling pathways in t & b cells..blocks proliferation of t cells and b cell proliferation and Ig synthesis
Definition
Describe action of Rapamycin
Term
elimination, equilibrium and escape
Definition
what are the three categories of cancer immunoediting
Term
represents the process of the immune system that selects or promotes cancer cells and initiates response of immune system to allow cancer to grow or to kill the cancer....important that this is the point that the immune response recognizes the tumor
Definition
Define equilibrium in cancer immunoediting
Term
mutated self protein overexpressed or abernathy expressed self protein oncogenic virus

important to denote here that tcells cd8 specifically is the type of cell involved in immunoediting
Definition
How are tumor cells displayihng what proteins/virus will be recognized by CD8 T cells
Term
loss/down regulation of HLA class I

down regulation, mutation or loss of tumor antigens

loss of co-stimulation alteration in cell death receptor signaling

immunosuppresive cytokines production of other suppressive factors
Definition
What are the 4 ways that cancer avoids immune surveillance?
Term
Stimulating the antitumor response

Decreasing suppressor mechanism

Altering tumor cells to increase their immunogenicity and make them more susceptible to immunologic defenses

Improving tolerance to cytotoxic drugs or radiotherapy
Definition
What are the main mechanisms of immunotherapy?
Term
antibody therapy

cytokine therapy

adoptive therapy-passively transfer cells to patient

vaccination

combinational therapy
Definition
What are some current immunotherapeutic strategies?
Term
Killed tumor cells

Purified tumor antigens

Dendritic cells pulsed with immunoglobulins

Plasmid DNA immunization
Definition
What are the types of tumor vaccines?
Term
Stimulating tumor cells by exposing them to tumor cells or antigens in the laboratory and the injecting expanded population of treated cells into the patient.

Patient is both donor and recipient
Definition
What is adoptive immunotherapy?
Term
blue
Definition
Color of staining for gram positive
Term
blue
Definition
Color of staining for acid fast gram positive
Term
pink
Definition
color of staining for gram-negative
Term
red or blue
Definition
color of staining for acid fast
Term
can't stain
Definition
color of staining for wall-less
Term

1-8 microns

 

coccus (cocci)

bacillus (bacilli)

spirochete

Definition
What are the main shapes of bacteria
Term
Peptidoglycan
Definition
What is the bacterial cell was composed of?
Term

40+

 

1-2

Definition
How many layers of peptidoglycan do gram-positive bacteria have?  Gram-negative?
Term
found in tears, saliva, and nasal secretions
Definition
What is a natural antimicrobial found in the human body that targets the peptidoglycan of bacteria?
Term

N-acetyl glucosamide - N-acetyl muramic acid

 

Have b(1,4) linkages

Definition
What are the monomers of peptidoglycan?
Term

L and D amino acids

 

Variability in these side chains are a typing mechanism for different species

Definition
What serves as linkages for side chains in peptidoglycan layers?
Term
Lipid A which is in the embedded in the outer membrane
Definition
What mediates septic shock in gram-negative bacteria?
Term

Peptidoglycans

 

40+ layers

Definition
Gram Positive cell walls are composed of what?
Term

Transformation

 

Transduction

 

Conjugation

Definition
Horizontal Exchange
Term

Meiosis

 

Zygote fusion

Definition
Vertical Exchange
Term

Gram Positive

 

Peptidoglycan layer is so thick that the MAC cannot penetrate the cell wall.

Definition
What type of bacterial cell is resistant to complement lysis?  Why?
Term

Waxes and Lipids

 

These hold onto the acid stains, which gives the cells their names.

Definition
What is the cell wall composed of in acid-fast bacteria?
Term

Flagella

 

capsule

 

pili

 

cell wall

 

endospore

Definition

What are the major components of the prokaryotic cell that can become virulence factors?

Term
Lipid A
Definition

Part of the outer membrane that mediates septic shock with a gram negative organism

Term

Thick Peptidoglycan Layer (40+)

 

Teichoic Acids and Lipoteichoic Acids

 

Other Proteins and Carbohydrates

Definition

Important Components for Gram Positive Bacteria

Term

Has an Outer Phospholipid Bilayer Membrane

 

Periplasmic Space

 

Single Layer of Peptidoglycan

 

Porins

 

Lipopolysaccharide (LPS)

Definition

Important Components for Gram Negative Bacteria

Term
The Periplasmic Space
Definition

Where do b-lactamases reside in the gram negative bacteria?

Term

b-lactamases are enzymes that break down b-lactams (penicillins).  Therefore, the organism is resistant to those microbials.

Definition

What is the importance of b-lactamases?

Term

Bacteria in the Bloodstream (Bacteremia) breaks down.  A component of the lipopolysaccharide, Lipid A, helps is engulfed or attached to a macrophage.  The macrophage, which then releases IL-1 and TNF-a, which work synergistically to induce fever, vasodilation, and “leaky” capillaries for chemotaxis of immune cells into tissue. This allows a severe drop in blood pressure and volume.  

Definition

What Is Happening During Septic Shock?

Term

Treatment centers around two key concepts:  dynamically addressing the primary pathogen responsible for the mediation into sepsis and reversing the hypersensitivity reaction within the individual.  Antibiotics such as tetracyclines have shown to be effective.  Surgical debriidement of infected tissue removes infected cells and bacterial mass.  However, this may lead to a susceptibility to an opportunistic pathogen.  Hemodynamic support, as in pushing fluids and vasopressor agents aid in the reversal of hypersensitivity reactions by the immune system.

Definition

What are accepted and developing methods for treating septic shock? 

Term

Lysozyme hydrolyzes the backbone and is naturally found in tears, saliva, and nasal secretions

Definition

Naturally occurring enzyme that breaks down the peptidoglycan and where it is found

Term

Main Chain is N-acetyl glucosamine and N-acetyl muramic acid (b1,4 linkage)

 

Cross-links are tetrapeptide with both L and D amino acids

Definition

Composition of the murein backbone (peptidoglycan)

Term

It is found in all species, though is highly variable between species.  It is used for typing.

Definition

What is significant about the tetrapeptide sequence in the murein backbone?

Term

At the b1,4 linkage between N-acetyl glucosamine and N-acetyl muramic acid

Definition

Where does lysozyme cleave the murein backbone?

Term

Smooth bacteria include the O-antigen on the end of their LPS, while rough end at the core.         

Definition

What is the difference between smooth and rough bacteria?

Term

Acid Fast Bacteria (Mycobacteria)

Definition

These bacteria have a cell wall that is composed of fatty acids and waxes that contribute to virulence.

Term
Capsule
Definition

K antigen

Term
Flagella
Definition
H Antigen
Term

 Filament (major straight structure that is swung around)

 

Hook (where we have the bend)

 

Rings (insertions into the membranes)

Definition

Main components of the flagella

Term

Pili

Definition

Hair-like proteins that are responsible for adhesion

Term

Conjugation

Definition

Sex pili are used in which process

Term

Fibular Layer

Definition

Protein coat on the surface of bacteria that can be a virulence factor

Term

Lag Phase (first introduction to environment – active metabolism)

 

Log Phase (rapid cell growth – population doubling every generation)

 

Stationary Phase (balance between reproduction and death – endospores are formed)

 

Death Phase

Definition

Phases of Bacterial Growth

Term

Bacteria that have 0-20°C  optimum growth.  No known psychrophilic human pathogens.

Definition

Psychrophiles 

Term

Bacteria that have 20-40°C preferred range, optimum 35-37°C = 98°F body temp (most human pathogens).  This causes special concerns with refrigerated foods

Definition

Mesophiles 

Term

Bacteria that have 40-90°C growth range

Definition
Thermophiles
Term

can tolerate ~2=15% salt

Definition

facultative halophiles 

Term

need up to 30% salt to grow

Definition

extreme halophiles 

Term

differentiates based on hemolysis reactions

Definition

Blood agar

Term

differentiates based on casease production

Definition

Milk agar

Term

differentiates based on type of lactose fermentation (mixed acid/neutral)

Definition

MRVP broth

Term

Differentiates based on lactose fermentation

Definition

MacConkey’s agar

Term

Normal Flora of Skin

Definition

Staphylococcus epidermidis & S. aureus

Propionibacterium, Corynebacterium

Pityosporum & Candida (fungi)

Term

Normal Flora of Upper Respiratory Tract

Definition

Staphylococcus epidermidis & S. aureus

Streptococcus pneumoniae, Hemophilus, Neisseria, diptheroids

Term

Normal Flora of Mouth

Definition

Streptococcus, Lactobacillus,

Corynebacterium Actinomycetes, Treponema,

Bacteroides, Fusobacterium, Candida

Term

Normal Flora of Large Intestine

Definition

Gram negative enterics (E. coli, Enterobacter, Citrobacter, Proteus, Klebsiella, Shigella), Enterococcus, Lactobacillus, Bacteroides, Fusobacterium, Clostridium, Candida

Term

Normal Flora of Urethra

Definition

S.epidermidis, Micrococcus, Enterococcus,

Lactobacillus, Pseudomonas, Klebsiella, Proteus, diptheroids

Term

Normal Flora of Vagina

Definition

Lactobacillus, Streptococcus, Staphylococcus,

Bacteroides, Clostridium, Candida, Trichomonas vaginalis (protozoan)

Term

Normal Flora of Male external genitalia

Definition

Mycobacterium smegmatis, other skin flora

Term

Damage to DNA

Protein Denaturation

Disruption of Cell Membrane of Wall

Removal of Sulfhydral Groups

Definition

Modes of Action for Antimicrobials

Term

Plasmid

Definition

Specialized Genetic Elements Capable of Independent Replication

Term

Transposons

Definition

Genetic Elements that Moves from One Gene Locus to Another

Term

Transformation

 

Conjugation

 

Transduction

Definition

Mechanisms of Bacterial Genetic Transfer

Term

Nt = N0 2n

where n is the number of generations

 

n = (log Nt – log N0)/0.301

Definition

Equation for Unchecked Binary Fission

Term
Generation Time
Definition

Time needed for a culture population to double

Term

1.      Microbe found in diseased animals and absent from healthy animals

 

2.      Microbes isolated and grown in culture

 

3.      Cultures causes disease in healthy animals

 

4.      Microbe is re-isolated from diseased animals and re-cultured

Definition

Koch’s Postulates

Term
ACME
Definition

Peak of Disease is known as 

Term

1.      Incubation period (no signs or symptoms)

2.      Prodromal phase (vague symptoms)

3.      Invasive Phase (most severe signs and symptoms)

4.      Decline Phase (declining signs and symptoms)

5.      Convalescence Period

Definition

Phases of Bacterial Infection

Term

Portals of Entry

Number of Invading Microbes

Adherence

Penetration or Evasion of Immune Defense

Cytopathic Effects

Portals of Exit

Definition

What are the Important Points of Transmission and Infectious Process

Term

Pili

Lipoteichoic Acid, F and M Proteins

Definition

Virulence Factors that Deal with Adherence

Term

Exotoxins are produced inside gram positive bacteria and are secreted or released into surrounding media.

 

Endotoxins are incorporated into the outer surface of the cell wall in gram negative bacteria.  They are liberated when the cell breaks apart.

Definition

What is the difference between Exotoxins and Endotoxins?

Term

Fragment A (inhibits protein synthesis)


Fragment B (binds to surface of CD by CD2)

Definition

What are the two fragments of Diptheria Toxin

Term

Protein A binds the Fc region of IgG

Definition

Why is Protein A expression on the surface of microbes considered an antiphagacytic agent?

Term

Purification of Antibodies

Definition

What is Protein A also used for?

Term

Genetic recombination presents a myriad of possible combinations for pili structure.

Definition

How do pili evade the immune system?

Term

Penicillians

 

cephalasporins

 

bacitracin

 

vancomycin

Definition

Antimicrobials that Inhibit Cell Wall Synthesis

Term

Quinolones

 

rifampin

Definition

Antimicrobials that Inhibit Nucleic Acid Replication and Transcription

Term

Chloramphenicol

 

erythromycin

 

tetracyclines

 

streptomycin

Definition

Antimicrobials that Inhibit Translation

Term

Sulfanilamides

 

trimethoprim

Definition
Antimicrobials that Inhibit Metabolite Synthesis
Term

Polymyxin B

Definition

Antimicrobials that Cause Injury to Plasma Membrane

Term
20-300 nm
Definition

Viral Size

Term

Capsid

Definition

Protein Shell That Coats And Encloses Nucleic Acid (RNA Or DNA)

Term

Capsomere

Definition

Make Up Capsid.  Morphologic Units (Polypeptides) 

Term

Classified as + or -.  Lipid containing membrane surrounding some viral particles. 

Becomes very important when we look at the release of some viral particles.

As the virus exits the cell, it may carry host membrane components.

Definition

Describe the Envelope of a Viral Particle

Term

Nucleocapsid

Definition

Protein/Nucleic Acid Complex 

Term

naked ssDNA viron with icosahedral symmetry that causes 5th disease

Definition

Parvoviridae

Term

enveloped dsDNA viron with icosahedral symmetry that causes HPV and genital warts

Definition

Papillomaviridae

Term

dsDNA viron with complex symmetry and coats that causes Herpes Type 1 and 2

Definition

Herpesviridae

Term

enveloped RNA viron that causes SARS

Definition

Coronaviridae

Term

enveloped RNA viron that causes HIV

Definition

Retoviridae

Term

enveloped RNA viron with helical symmetry that causes flu

Definition

Orthomyxoviridae

Term

enveloped RNA viron with helical symmetry that causes rabies

Definition

Rhabdoviridae

Term

Attachment

Penetration

Uncoating

Expression of viral genome

Synthesis of viral components

Assembly

Release

Definition

Basic Steps for Viral Replication Cycle

Term

Causes Mononucleosis

 

Attaches via CD21 on B cells

Definition

Epstein-Barr Virus

Term

Respiratory

 

Most exposure to the environment

Definition

The most common port of entry for viral infections is

Term

Cilia, lymphocytes, macrophages, IgA

Definition

Localized Immune Response to Respiratory Viral Infections

Term
33.2 million
Definition
How many cases worldwide do we have of HIV?
Term
2.5 million
Definition
Worldwide New Infection Rate with HIV for 2007
Term

Sub-Saharan Africa

 

22.5 million cases

 

Especially in Zimbabwe and Botswana

Definition
Where is the heaviest accumulation of HIV cases seen?
Term
HIV-1 is a lentivirus
Definition
What strain of HIV and what type of virus is responsible for causing AIDS?
Term

Binding and Entry

 

Reverse Transcription

 

Replication

 

Budding

 

Maturation

Definition
Phases of the HIV Life Cycle
Term

When the CD4 cell count falls below 200 cells/ml

 

and/or

 

A typical AIDS-defining illness is observed

Definition
When is an HIV infection considered AIDS?
Term
viral load
Definition
Which presents a good correlation of disease progression with HIV, viral load or CD4 cell count?
Term
CCR5, CCR2, and CXCR4
Definition
What are "cofactors" for HIV infection?
Term

Erythema infectisum.

Mild constitutional symptoms may accompany the rash, which has a typical “slapped cheek” appearance.  Join involvement is a prominent feature in adult cases.  The symptoms mimic RA.

Definition

Clinical manifestation of parvovirus (Fifth Disease)

Term

Treated symptomatically

Definition

Treatment for Fifth Disease

Term

Oropharyngeal:  HSV-1 involves the buccal and gingival mucosa of the mouth.  Symptoms include fever, sore throat, vesicular and ulcerative lesions, gingivostomatitis, and malaise.

Keratoconjunctivitis: HSV-1 may occur in the eye, producing sever keratoconjunctivtis

Genital Herpes: HSV-2 causes genital herpes is characterized by vesiculoulcertive lesions of the penis of the male or the cervic, vulva, vagina, and perineum of the female

Definition

Clinical manifestations of Herpesviruses

Term

Several antiviral drugs have proved effective against HSV infections, including acyclovir, valacyclovir, and vidarabine.  Acyclovir is currently the standard therapy. 

All are inhibitors of viral DNA synthesis.

Definition

Treatment of Herpes

Term

Symptoms of classic influenza usually appear abruptly and include chills, headache, and dry cough, followed closely by high fever, generalized muscular aches, malaise, and anorexia.

Definition

Clinical manifestation of orthomyxovirus (influenza)

Term

The NA inhibitors zanamivir and oseltamivir were approved in 1999 for treatment of both influenza A and B. 

To be maximally effective, the drugs must be administered very early in the disease.

Definition

Treatment of Influenza

Term

The human coronavirses produce “colds” usually afebrile in adults.  The tymptoms are similar to those produced by rhinovirses, typified by nasal discharge and malaise. 

Symptoms usually last about 1 week.

Definition

Clinical manifestation of coronavirus (SARS)

Term

 

There is no proven treatment and no vaccine.

 

Definition

 

Treatment for SARS

 

Term

 

The clinical spectrum can be divided into three phases

a short prodromal phase – malaise, anorexia, headache, photophobia, nausea, sore throat and/or fever

acute neurologic phase – signs of nervous system dysfunction

coma phase

 

Definition

 

Clinical manifestation of rabies

 

Term

 

Prevention by Vaccine

 

Definition

 

Treatment of Rabies

 

Term

Source: Macrophages (IFN-a), fibroblasts (IFN-b)

 

Function: Antiviral

Definition
IFN-a, -b
Term

Source: T Cells and NK Cells

 

Function: Activateion of macrophage and TH1 differentiation

Definition
IFN-g (interferon)
Term

Source:  Macrophages and T cells

 

Function:  Cell activation, fever, cachexia, antitumor

Definition
TNF-a
Term

Source: T cells

 

Function:  Activates PMNs

Definition

TNF-b

Term

Source:  Macrophages

 

Function:  Cell activation, fever

Definition
IL-1
Term

Source:  T cells

 

Function:  T cell growth and activation

Definition
IL-2
Term

Source: T cells and mast cells

 

Function: B cell proliferation and switching to IgE

TH2 differentiation

Definition
IL-4
Term

Source: Macrophages

 

Function: Differentiation of T cells

activation of NK cells

Definition
IL-12
Term

Spores germinate in the tissue at the site of entry, and growth of the vegetative organisms results in formation of gelatinous edema and congestion.  Bacilli spread via lymphatics to bloodstream shortly before and after death.

Definition

Characteristics of spores of Bacillus anthracis

Term

Capsule is required for virulence.  The poly-D-glutamic acid capsule is antiphagocytic.  The capsule gene is on a plasmid.

Definition

Characteristics of capsule of Bacillus anthracis

Term

Anthrax toxin is made up of three proteins: protective antigen (PA), edema factor (EF), and lethal factor (LF).  PA binds to specific cell receptors, and following proteolytic activation it forms a membrane channel that mediates entry of EF and LF into the cell.

EF is an adenylyl cyclase; with PA it forms a toxin known as edema toxin. 

LF plus PA form lethal toxin, which is a major virulence factor and cause of death.

Definition

Characteristics of Anthrax toxin from Bacillus anthracis

Term

Many antibiotics are effective against anthrax in humans, but treatment must be started early. 

Ciprofloxacin is recommended for treatment; penicillin G, along with gentamicin or streptomycin, has previously been used to treat anthrax.

Definition

Treatment of infection with Bacillus anthracis

Term

Food poisoning caused by Bacillus cereus has two distinct forms

The emetic type is associated with fried rice

The diarrheal type is associated with meat dished and sauces.

Definition

What is an infection of Bacillus cereus associated with?

Term

Nausea, vomiting, abdominal cramps, and occasionally diarrhea.  Recovery occurring within 24 hours.

Definition

Clinical manifestation of the emetic form of B cereus infection

Term

Incubation period of 1-24 hours is followed by profuse diarrhea with abdominal pain and cramps.  Fever and vomiting are uncommon.

Definition

Clinical manifestation of diarrheal form of B cereus infection

Term

An exotoxin that has seven antigenic variants, however types A, B, and E are the principal causes of human illness.  Types A and B have been associated with a variety of foods and type E predominately with fish products. 

The toxin is a large protein that is cleaved into two proteins linked by a disulfide bond.

Definition

Characteristics of Clostridium botulinum toxin

Term

Toxin is absorbed from the gut and binds to receptors of presynaptic membranes of motor neurons of the PNS and cranial nerves. 

After cleaving of the toxin, it inhibits the release of ACh at the synapse, resulting in lack of muscle contraction and paralysis.  

Definition

Pathology of Clostridium botulinum toxin

Term

Yes, the toxins are destroyed by heating for 20 minutes at 100 °C.

Definition

Can Clostridum botulinum toxin be destroyed?

Term

Visual disturbances (incoordination of eye muscles), inability to swallow, and speech difficulty, signs of bulbar paralysis are progressive, and death occus from respiratory paralysis or cardiac arrest.

Definition

Clinical manifestation of oral digestion of Clostridum botulinum

Term

Potent antitoxins to three types of botulinum toxins have been prepared in horses.  Since the type responsible for an individual case is not known, trivalent antitoxin must be promptly administered with customary precautions.

Definition

Treatment of Clostridum botulinum

Term

The toxin initially binds to receptors on presynaptic membranes of motor neurons, and migrates to the CNS.  The toxin diffuses to terminals of inhibitory neurons, and degrades a protein required for docking of neurotransmitter vesicles on the presynaptic membrane.  Release of the inhibitory neurotransmitters is blocked, and motor neurons are not inhibited.  Spastic paralysis results.

Definition

Pathology of Clostridum tetani toxin

Term

The disease is characterized by tonic contraction of voluntary muscles (lockjaw).  Gradually, other moluntary muscles become involved, resulting in tonic spasms.  Any external stimulus may precipitate a titanic generalized muscle spasm.  Death usually results from interference with the mechanics of respiration.

Definition

Clinical manifestation of Clostridium tetani

Term

The IM administration of human antitoxin give adequate systemic protection.  It neutralizes the toxin that HAS NOT BEEN FIXED to nervous tissue.

Patients who develop symptoms are treated symptomatically, with muscle relaxants, sedation, and assisted ventilation.  Penicillin strongly inhibits the growth of bacteria and prevents further toxin production.

Definition

 

Treatment of Clostridium tetani

 

Term

Closteridium perfringens

Definition

Bacteria that is the causal agent for gas gangrene

Term

The alpha toxin is a lecithinase, and its lethal action is proportionate to the rate of splitting.  The theta toxin has similar hemolytic and ncrotizing effects but is not a leithinase.  DNase and hyaluronidase are also produced.

Definition

Characteristics of Clostridium perfringens toxin

Term

From a contaminated wound, production of a foul smelling discharge, rapidly progressing necrosis, fever, hemolysis, toxemia, shock, and death. 

Food poisoning usually follows the ingestion of large numbers of this bacteria  that have grown in warmed meat dishes.  The toxin forms when the organisms sporulate in the gut, with the onset of diarrhea – usually without vomiting or fever. 

Definition

Clinical manifestation of Clostrdium perfringens

Term

Administration of antimicrobial drugs, particularly penicillin, is begun at the same time as surgical debridement if cutaneous.  Food poisoning due to entertoxin usually requires only symptomatic care.

Definition

Treatment of infection with Clostridium perfringens

Term

There are over 35 different species, three of which are of clinical significance.

Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus saprophyticus

Definition

The genus Staphylococcus has how many species?  How many and which ones are of clinical significance

Term

Catalase

Definition

What is an enzyme that is produced that differentiates staphylococci from streptococci?

Term

Catalase – converts H2O2 into H2O and O2

 

Coagulase – initiates fibrin polymerization

 

Clumping factor – responsible for adherence of organisms to fibrinogen and fibrin

 

Exotoxins – attacks membranes (cell membrane and sphingomyelin)

 

Leukocidin – attacks and kills white blood cells

 

Exfoliative toxins – superantigens that cause desquamation

 

Toxic Shock Syndrome Toxin – TSST-1 binds to MHC class II which promotes toxic shock symptoms

 

Enterotoxins - superantigens

 

Definition

 

What are the main substances and toxins that are associated with staphylococci?

 

Term
Streptococcus pyogenes
Definition
Most bacteria in Group A Streptococci (GAS)
Term
Beta
Definition
Type of Hemolysis with S pyogenes
Term

Pharngitis

 

Impetigo

 

Rheumatic Fever

 

Scarlet Fever

 

Glomerulonephritis

Definition
Common and Important Diseases associated with S. pyogenes (GAS)
Term

Throat

 

Skin

Definition
Habitat of S. pyogenes
Term

Composed of hyaluronic acid

 

Impede phagocytosis

Definition
Describe the capsule of GAS
Term

Consist partly of M protein

 

Covered with lipoteichoic acid (important for attachement to EC)

Definition
Describe the pili of GAS
Term

Streptokinase (fibrinolysin to break up clots)

 

Streptodornase (depolymerizes DNA)

 

Hyaluronidase (spreading factor)

 

Pyrogenic Exotoxins (Erythrogenic Toxin - associated with Streptococcal toxic shock syndrom and scarlet fever)

 

Diphosphopyridine Nucleotidase (aid in killing leukocytes)

 

Hemolysins

Definition
What are the important Toxins and Enzymes produced by S. pyogenes
Term

pharyngitis

 

Pili attache to epithelium via lipoteichnoic acid. 

 

In infants and small children, the sore throat occrs with little fever and extends to middle ear and the mastoid.  Cervical lymph nodes are typically enlarged. 

 

In older children and adults, disease is more acute.  Characterized by intense nasopharyngitis, tonsilitis, and intense redness and edema of the mucous membranes and high fever.

Definition
Name and describe the most common infection due to S pyogenes
Term

Consists of superficial vesicles that break down and erode areas where surface is covered with pus and later is encrusted.  It spreads continuity and is highly communicable. 

 

More widespread infection occurs in eczematous or wounded skin or in burns and may progress to cellulitis.

Definition
Describe impetigo caused by S pyogenes
Term

Penicillin G

 

most are also susceptible to erythromycin

Definition
Treatment of choice for S pyogenes
Term
Penicillin's action is to inhibit cell wall growth (formation of peptidoglycan cross-links)
Definition
What is the mode of action for the first antibiotic of choice for GAS?
Term
False.  Antigens may be present with killed bacteria.
Definition
T/F:  A positive antigen test for GAS, indicates the presence of a live bacteria.
Term
Urinary tract
Definition
Predominant Site for E coli infections
Term
staphylococci and streptococci
Definition
Most common Enterobacteriacae causing GI inflammation
Term

Gram negative rods

 

many times are motile due to peritrichous flagella

 

may or may not have a capsule

 

may be aerobic or anaerobic

Definition
Describe the general class of Enterobacteriacae
Term

Lipopolysaccharide side chains

 

Over 150 O serotypes

Definition
O antigens
Term

Capsule

 

Over 100 capsular antigens

Definition
K antigens
Term

Flagella

 

Over 50 flagellar antigens

Definition
H antigen
Term

E coli causes 90% of UTIs

 

K antigen (capsule) is mainly responsible

Definition
What is the most common cause of urinary tract infection, what percentage of UTIs are caused this way, and what it the primary virulance factor used?
Term

EPEC is an important cause of diarrhea in infants.  Nosocomial and community infection for developed countries.

 

Adhere to mucosal cells of bowls by chromosomal encoded adherence factors.  Loss of microvilli and formation of pedestals.

 

Result is self-limiting, watery diarrhea.

 

Duration shortened and cured by antibiotic treatment. 

Definition
Discuss Enteropathogenic E coli
Term

ETEC is a causitive agent for "traveler's diarrhea" or "Montizuma's Revenge" and diarrhea for infants in developing countries.

 

Produce toxins LT and ST (discussed on another card)

 

Spread by fecal contamination.

 

At least 108 E coli need to be present to result in transmission.

Definition
Discuss Enterotoxigenic E coli
Term

Organisms produce LT and/or ST

 

LT (heat-labile exotoxin) is a high MW protein in two subunits.  Subunit B attaches to EC and facilitates entry of Subunit A.  Hypersecretion of water and chlorides results, with the inhibition of reabsorption of sodium.

 

ST (heat-stable enterotoxin) is a low MW protein that stimulates fluid secretion.  

 

Both toxins are found on a plasmid.

Definition
Describe the toxins produced by ETEC
Term

EHEC is a causitive agent for hemorrhagic colitis ("Hamburger disease") with hemolytic uremic syndrome.  Causes DIC in arterioles and arteries of nephrons, decreasing kidney function and potentially acute kidney failure and thrombocytopenia.  

 

Extremely low innoculum required for disease.  Most common serotype is O157:H7.

 

Many cases can be prevented by thoroughly cooking ground beef.

Definition
Discuss Enterohaemorrhagic E coli
Term

EIEC is similar to shigellosis

 

Invasion of epithelial cells

 

Food and water borne.

 

Most common in children in developing countries.

Definition
Discuss Enteroinvasive E coli
Term

Shigellae are slender gram-negative rods, that are aerobic or anaeorobic, and ferment glucose.  Shigella sonnei also ferments lactose.

 

More than 40 O serotypes

Definition
Describe the organism of shigelae
Term

Endotoxins (lipid A)

 

Shigella dysenteriae exotoxin

Definition
Two types of toxins from Shigellae
Term

Heat-labile exotoxin that affects both the gut and CNS.  The exotoxin is a protein that is antigentic, thus may illicit antitoxin production.

 

Produces diarrhea and inhibits sugar and amino acid absorption in the small intestine, proceeds to dysentery with blood and pus in stools.  The neurotoxin effect may rsult in meningismus or coma.

Definition
Describe the Shigella Dysenteriae Exotoxin
Term

Motile organisms with peritrichous flagella.  The classification of salmonellae is complex because the organisms are a continuum rather than a defined species.  Antigenic structures O, H, and Vi (equivalent to K) are important pathogenic factors.  

 

The organisms enter via the oral route, usually with contaminated food or drink.  Host factors contribute to resistance.  Salmonellae produce three main types of disease in humans (enteric fevers, septicemias, and enterocolitis), but mixed forms are frequent.

Definition
Describe Salmonella
Term

Produced by only a few of the salmonellae, of which Salmonella typhi is the most important.  

 

Ingested salmonellae reach the small intestine, go through the lymphatics to the blood stream.  The organisms multiply in the intestinal lymphoid tissue and are excreted in stools.

 

After an incubation period of 10-14 days, fever, malaise, headache, constipation, bradycardia, and myalgia occur.  A gradual rise then high plateau in fever occurs.

 

Antimicrobials have reduced mortality rate from 10-15% to less than 1%.

Definition
Describe Enteric Fever (Typhoid Fever)
Term

Most common manifestation of salmonella infection.  Organism penetrates and replicates inside of cell.

 

8-48 hours after ingestion, nausea, headache, vomiting, profuse diarrhea with low-grade fever occurs.  Episode usually resolves in 2-3 days.  

 

Bacteremia is rate (2-4%).

Definition
Describe Enterocolitis infection from Salmonella species
Term

Abrupt onset of liquid diarrhea in edemic area, up to 5 L/day in severe forms.  Needs prompt replacement of fluids and electrolytes IV or orally.  Killer in 3rd world countries.

 

Ogranisms gow in gut and produce toxin (describe on another card).  Toxin causes hypersecretion in small intestine.  Disease is toxin-mediated.  

Definition
Describe clinical features, epidemiology and pathogenesis of Vibrio cholarae
Term

  1. Permenant activation of G protein
  2. Adenylene cyclase activated to produce 2nd messenger cAMP
  3. Release of potassium, chlorine, sodium, and bicarbonate ions.
  4. Change in osmotic pressure leads to diarrhea

Definition
Describe the process invoked by Cholera Toxin
Term

Fever, diarrhea; PMNs and fresh blood in stools, especially in children.  Usually self-limited.

 

Infection via oral route from food and pets.  Organisms grow in the small intestine.  Invasion of mucous membrane.  Any toxins are uncertain.

Definition

Describe clinical features, epidemiology and pathogenesis of Campylobacter jejuni

 

Term

Upper gastrointestinal illness with nausea and pain.  About 90% of patients with duodenal ulcers and 50-80% of those with gastric ulcers are caused by H pylori.  

 

Spiral-shaped gram negative rod with many flagella on one end that grows best in pH 6-7 at 37 °C.  

 

H pylori is found deep in the mucous layer where the pH is more suitable for growth.  The bacterium overlies gastric-type EC.  Production of a protease that modifies the gastric mucus, reduces the ability of acid to diffuse through the mucus.  Potent urease activity, which yields production of ammonia and further buffering of acid.

Definition

Describe clinical features, epidemiology and pathogenesis of Helicobacter pylori

Term

Nomenclature confusion as this is not a virus, but a gram negative rod, with capsular polysaccharide.

 

No exotoxin, and causes flu-like symptoms.  Therefore many times is mis-diagnosed as the seasonal flu.

 

High carrier-rate

Definition
Describe Haemophilus influenzae
Term

Small gram-negative rod that casues chancroid (soft chancre), an STD.

 

Ragged ulcer on the genitaliaz.

 

Treatment with IM ceftriaxone, oral Bactrim, or oral Erythromycin.

Definition
Describe Haemophilus ducreyi
Term

Gram-negative coccobacilli that has an vaccine (though boosters are needed)

 

Organisms colonize epithelium after transmission via the respiratory route.  After a 2 week incubation, the "catarrhal stage" develops, shere large numbers of organisms are sprayed in droplets during mild coughing.  During the "paroxysmal" stage, the cough develops its explosive character and the characteristic "whoop" upon inhalation.

 

Toxins

  • Pertussis toxin - causes cough
  • Adenylate cyclase toxin - damage to cell signaling
  • Dermonecrotic toxin - necrotic condition of skin
  • Hemolysin - irritation of bronchiols that contributes to cough and secondary infections

 

 

Definition
Describe Bordetella pertussis
Term

Abrupt onset of fever

 

Hypotension

 

Headache

 

Stiff neck (nuchal rigidity)

 

Rash

Definition
Clinical manefestations of Neisseria meningitidis
Term

Transmission is via respiratory droplets

 

The nasopharynx is the portal of entry, however can become a transient portion of the flora.

 

Temporal pattern peaks in late winter to early spring.

Definition
Transmission and Portal of Entry for Neisseria meningitidis
Term

Aerobic gram-negative bacteria

 

At least 13 serogroups based on polysaccharide capsule

 

Relative importance of serogroups depends on geographic location and other factors, such as age.

Definition
Describe the organism of Neisseria meningitidis
Supporting users have an ad free experience!