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

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/490

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

490 Cards in this Set

  • Front
  • Back
Complete deficiencies of which molecules are not compatible with life?
Hb alpha (Hydrops fetalis, --/--)
Protein C
Protein S
Antithrombin
Prothrombin
Tissue factor
1. How does TF activate coagulation in vitro and in vivo?
2. What protein is responsible for this discrepancy?
1. In vitro: TF converts VII-->VIIa, VIIa converts X-->Xa
In vivo: TF converts IX-->IXa, IXa+VIIIa converts X-->Xa

2. TFPI (tissue factor pathway inhibotr) binds/inhibits Xa and then binds/inhibits VIIa:TF
1. Which molecules are Vitamin K dependent?
2. Which has the shortest half-life?
1. II
VII
IX
X
Protein C
Protein S

2. VII
Why would an antibody to TFPI cure an animal model with hemophilia A or B?
Hemophilia A (VIII deficiency)
Hemophilia B (IX deficiency)
Intrinsic pathway cannot activate
Antibody to TFPI allows VII-->VIIa:TF and X-->Xa in vivo (like PT in vitro)
1. Which three molecules does antithrombin inactivate?
2. What is the difference in the mechanisms of action of low and high MW versions of heparin and heparin-like proteoglycans?
1. Thrombin, Xa, IXa

2. Low MW heparins + Antithrombin --> Xa inactivation
High MW heparins + Antithrombin --> Thrombin, Xa, IXa inactivation
What are the different actions/states of Plasmin, alpha-2-PI, and PAI-1 when in the plasma versus in a fibrin clot?
Plasmin binds to lysine residues of fibrin and degrades fibrin. Free circulating plasmin degrades Va and VIIIa, decreasing fibrin formation.

Alpha-2-PI is covalently linked to fibrin by XIIIa and inhibits plasmin. Free Alpha-2-PI is rapidly degraded, increasing fibrinolytic activity.

PAI-1 is bound to subendothelial vitronectin or secreted where tPA is actively secreted, and it inhibits tPA, inhibiting overall fibrinolysis. Free PAI-1 is rapidly degraded, increasing fibrinolytic activity.
1. How is TAFI activated?
2. What is TAFI's function?
3. What is TAFI's effect?
1. Thrombin+Thrombomodulin activates TAFI.
2. TAFI removes terminal lysine and arginine residues, which are required for tPA and plasminogen to bind.
3. TAFI inhibits the generation and activity of plasmin, thus decreasing fibrinolysis.
What does the protamine paracoagulation test test for?
Soluble fibrin monomer
What are the 6 major causes of disseminated DIC?
Infection
Obstetrical complications
Neoplastic disease
-PML
-Disseminated neoplasm
Massive tissue necrosis
Prolonged shock
Miscellaneous
What are the 5 major causes of localized DIC?
Kasabach-Merritt Syndrome
Phlegmasia Cerulean Doleans
Aortic aneurysm
Hyperacute renal transplant rejection
Miscellaneous
DIC is characterized by which form of fibrinolysis?
Secondary fibrinolysis
What are the 4 consequences of DIC?
Bleeding
Ischemic tissue damage
Stroke/acute MI
Fragmentation hemolytic anemia
What does cryoprecipitate contain?
VIII
Fibrinogen
vWF
XIII
1. Aside from curing the underlying cause, what is the standard treatment for bleeding and consumption of coagulation factors caused by DIC?
2. When is heparin indicated for DIC management?
3. When is heparin contraindicated for DIC management?
If bleeding and significant depletion of clotting factors and/or platelets are clinical problems, FFP or cryoprecipitate should be given.

Heparin indications:
-Long term DIC causing poor response to replacement therapy
-Thrombosis or ischemia

Heparin contraindications:
-Easily removed or short-lived DIC
-No bleeding or thrombotic complications
-Bleeding easily controlled by factor replacement
What are the characteristics of arterial thrombi?
High shear conditions
Platelet and vWF dependent
Associated with atherosclerosis
Often occurring at branch points
What are the characteristics of venous thrombi?
Low shear conditions
Thrombin and fibrinogen dependent
Minimally associated with atherosclerosis
What substrates does platelet GpIIb/IIIa bind to, and under what conditions does the binding occur?
High shear (arteries)
vWF:GpIIb/IIIa

Low shear (veins)
Fibrinogen:GpIIb/IIIa
Which hemostatic molecules play a role in normal and pathologic angiogenesis? How?
TF:VIIa signals via PAR2, inducing VEGF production.
Thrombin signals via PAR4, inducing VEGF, IL-8, and IL-6 production.
What is the primary method of inhibiting venous thrombus formation?
Inhibition of thrombin formation
What is the mechanism of action of Hirudin and Bivalirudin?
Thrombin inhibition
Which drugs are used to inhibit thrombin formation as prophylaxis or treatment of thrombi?
Hirudin
Bivalirudin
What is the mechanism of action of thienopyridines (Ticlopidine, Clopidogrel)?
ADP-Receptor/P2Y<sub>12</sub> antagonism
Which drugs are used to antagonize ADP-Receptors/P2Y<sub>12</sub>?
Thienopyridines
-Ticlopidine
-Clopidogrel
What is the mechanism of action of Abciximab, Eptifibatide, and Tirofiban?
GpIIb/IIIa aggregation inhibition
Which drugs are used to inhibit GpIIb/IIIa aggregation?
Abciximab
Eptifibatide
Tirofiban
What dose of aspirin is required for antithrombotic effect?
<u>></u>75mg
Which drugs will mimic Glanzman's Thrombasthenia?
GpIIb/IIIa receptor antagonists:
Abciximab (irreversible)
Eptifibatide, Tirofibam (reversible)
Where do the stages of T cell maturation occur?
What are the stages of T cell maturation?
Thymic cortex: Positive selection
-ensures T cells can recognize MHCII/I:Peptide complexes with their TCR and CD4/8
Thymic medulla: negative selection
-Medullary epithelial cells transcribe the aire locus and express self-antigens to destroy self-reactive T cells
How do T<sub>reg</sub> cells arise?
Some self-reactive T cells in the thymic medulla bypass negative selection and become T<sub>reg</sub> cells (CD4<sup>+</sup>CD25<sup>+</sup>)
Where do the stages of B cell maturation occur?
What are the stages of B cell maturation?
Bone marrow: positive selection
-heavy then light chain rearrangements in the bone marrow are tested for efficacy
Bone marrow: Negative selection
-B cells which bind strongly to expressed self-antigen rearrange their light chains or are clonally deleted
How is T cell anergy and peripheral tolerance developed?
T cells in the periphery recognize self-antigens on APCs in peripheral tissues, but the T cells in the periperhy do not receive costimulatory signals from the APC or receive inhibitory signals from the APC B7 binding to the T cell CTLA-4
Explain activation-induced death of T lymphocytes
Repeated stimulation of activated T cells by self-antigen without the APC co-stimulatory signal leadss to a brief period of cell division, expression of Fas and FasL, and apoptosis
Explain T<sub>reg</sub> inhibition of T lymphocytes
T<sub>reg</sub> cells expressing CD4 and CD25 recognize self-antigen and indirectly or directly inhibit other T cells by releasing inhibitory cytokines or through inhibitory physical mechanisms
How is B cell anergy and peripheral tolerance developed?
B cells in the periphery which bind strongly to soluble self antigen decrease their expression of IgM (but retain normal IgD expression) and become inactive until they die
Hereditary deficiencies or dysfunctions in phagocytic cells leads to what pathologies?
Recurrent skin, lymph node, or deep tissue infections with
<i>S. aureus</i>
gm(-) (<i>Serratia spp.</i>)
fungi (<i>Candida albicans</i>, <i>Aspergillus spp.</i>)
Hepatosplenomegaly, lymphadenopathy
What causes CGD?
What are the associated pathologies?
X-linked recessive deficiency of Phagocyte NADPH Oxidase (<i>Phox</i> gene) leads to ineffective destruction of phagocytosed microbes and recurrent infections with <i>S. aureus</i>, gm(-) bacteria, and fungi.
Dx: NBT test for Phox activity
What is the mechanism of increased recurrent infections of those with phagocytic cell deficiencies?
Ubiquitous organisms (<i>S. aureus</i>, gm(-)'s, and fungi) do not require opsonizing antibodies for clearance, but the innate response is essential and, in this case, impaired.
Hyposplenism leads to what pathologies?
Recurrent bacterial sepsis due to encapsulated organisms (<i>S. pneumoniae</i>, <i>H. influenzae</i> type B)
What is the mechanism of recurrent bacterial sepsis due to encapsulated organisms in hyposplenic patients?
The spleen normally produces very rapid, natural, opsonizing antibodies which are necessary for clearance of <i>S. pneumoniae</i> and <i>H. influenzae</i>
What causes DiGeorge Syndrome?
What is the mechanism of immune dysfunction?
Chromosome 22 deletion causes varying T cell deficiencies (T cell maturation and signalling, deficient T cell-mediated responses, and deficient B cell help) ranging from asymptomatic, mild T-lymphocytopenia to thymic agenesis with a complete absence of T cells
What is the clinical phenotype of DiGeorge syndrome?
Viral, fungal, mycobacterial infections by opportunistic pathogens
GI symptoms
Oral candiasis
Hepatosplenomegaly/lymphadenopathy
Severe seborrhea-like dermatitis
Variable craniofacial dysmorphisms, cardiovascular conotruncal malformations, parathyroid hypocalcemia, and T lymphocytopenia
What is the clinical phenotype of B cell deficiency?
Upper/lower respiratory tract infections with the usual pathogens
Gi symptoms
Granulocytopenia or JRA-like arthritis
What is the mechanism of the clinical phenotype of B cell deficiency?
Mutations in B cell maturation and signalling lead to local or systemic deficient ag-specific antibodies required for efficient opsonization of encapsulated
What is the cause of X-linked agammaglobulinemia?
What are the characteristics of the syndrome?
Defective Btk on the X-chromosome results in a maturation block after the pre-B cell stage.
X-linked agammaglobulinemia is characterized by a complete lack of mature B cells, absent IgM and IgA antibodies, and normal T cell count and function.
What is the most common primary immune deficiency?
Selective IgA deficiency
What is the likely cause of Selective IgA deficiency?
What are the characteristics of the syndrome?
Variable genetic patterns cause a signalling defect resulting in failure of IgA isotype class-switching.
Selective IgA deficiency is characterized by <5mg/dL of serum IgA and undetectable levels of secretory IgA
What is the clinical phenotype of Selective IgA Deficiency?
What is the mechanism of this phenotype?
A lack of IgA can be asymptomatic or cause an increase in compensatory mechanisms, leading to atopic disease, recurrent infections, and auto-immune disorders
What is the cause of CVID?
What are the characteristics of CVID?
Variable genetic mutations can cause CVID.
CVID is characterized by variable hypoglobulinemia, age of onset, and degree of T cell abnormalities.
What is the clinical phenotype of CVID?
What is the mechanism of this phenotype?
CVID presents with recurrent bacterial infections due to hypogammaglobulinemia and impaired Ag-specific antibody responses, but a normal number of B cells.
Deficiencies in C5-C9 lead to susceptibility to which organism?
<i>Neisseria spp.</i>
Deficiencies in IFN-g lead to susceptibility to which organism?
Atypical mycobacteria
Deficiencies in TLR3 lead to susceptibility to which organism?
Herpes simplex encephalitis
What is the mechanism and phenotype of Grave's Disease?
Anti-TSH Receptor antibodies (agonism) cause hyperthyroidism
What is the mechanism and phenotype of Myasthenia Gravis?
Anti-ACh Receptor antibodies (antagonism) cause muscle weakness
What is the mechanism and phenotype of Insulin-Resistant Diabetes?
Anti-Insulin Receptor antibodies (antagonism) cause hyperglycemia and ketoacidosis
What is the mechanism and phenotype of Autoimmune Hypoglycemia?
Anti-Insulin Receptor antibodies (agonism) cause hypoglycemia
What is the mechanism and phenotype of Insulin-Dependent Diabetes Mellitus?
Effector T Cells target beta-antigen on beta cells of the pancreas, causing destruction of beta cells and the ability to produce insulin
What is the mechanism and phenotype of Rheumatoid Arthritis?
Effector T Cells target an unknown synovial joint antigen, causing destruction and inflammation of joints
What is the mechanism and phenotype of Multiple Sclerosis?
Effector T Cells target myelin basic protein, causing destruction of myelin, brain damage, and paralysis
Mutations in Fas and FasL lead to which autoimmune disorder?
What is the mechanism of pathology?
Fas, FasL
ALPS (Autoimmune Lymphoproliferative Disorder)
Defective elimination of self-reactive T and B cells by AICD (Activation Induced Cell Death)
Mutations in AIRE lead to which autoimmune disorder?
What is the mechanism of pathology?
AIRE
Autoimmune Polyendocrinopathy with Candidiasis and Ectodermal Dysplasia
Defective elimination of self-reactive T cells in the thymus due to an inability for negative selection
Mutations in FoxP3 lead to which autoimmune disorder?
What is the mechanism of pathology?
Foxp3
Immune dysfunction polyendocrinopathy enteropathy/X-linked
Defective T<sub>reg</sub> Cells do not downregulate Effector T Cells
Mutations in C2 and C4 lead to which autoimmune disorder?
What is the mechanism of pathology?
C2, C4
Lupus-Like Disease
Defective clearance of immune complexes
The HLA-B27 allele leads to an increase risk of what autoimmune disorder?
What is the mechanism of pathology?
HLA-B27
Ankylosing Spondylitis
Inflammation of the spine results in fusion and rigidity
Infection with EBV is associated with what autoimmune disorder?
Multiple Sclerosis
Infection with Group A Strep (<i>Streptococcus pneumoniae</i>) is associated with what autoimmune disorder?
Acute rheumatic fever
Acute glomerulonephritis
What is the most effective treatment for Multiple Sclerosis?
Natalizumab
MAB to alpha-4-integrin
What situations can lead to non-clonal thrombocytosis?
Iron deficiency
Chronic inflammation
1. What is polycythemia vera?
2. What are its CBC findings?
1. Accelerated normoblast maturation and differentiation into RBCs.
2. Drastically increased Hgb or Hct levels.
1. What is essential thrombocythaemia?
2. What are its diagnostic criteria?
1. Megakaryocyte proliferation and differentiation.
2. Platelet count <u>></u>450x10<sup>9</sup>
What is primary myelofibrosis?
Megakaryocyte proliferation and atypia accompanied by reticulin and/or collagen fibrosis.
1. What is the most common myeloproliferative disease?
Chronic Myeloid Leukemia (CML)
1. What chromosome abberation is associated with CML?
2. What is the protein product of the chromosome abnormality and its function?
1. Philadelphia Chromosome t(9;22)
2. bcr-abl fusion protein has constitutive tyrosine kinase activity causing increased cell division and DNA repair inhibition
What is the clinical presentation of chronic phase CML?
What is the clinical presentation of blast phase CML?
Chronic phase: Splenomegaly with elevated WBC count, with neutrophils from all maturation stages
Blast phase: Transformation to acute leukemia (70% myeloid, 30% lymphoid), anemia, thrombocytopenia, lymphoblasts or myeloblasts
How do we know that the HSC is involved in CML?
Chronic phase CML can progress to blast phase AML (70%) or ALL (30%), indicating that the progenitor to both cell lines is affected.
How do patients die from CML?
Increased proliferation and accumulation of mutation leads to an acute blastic crisis.
What is the cure for CML?
Allogeneic stem cell transplant
1. What is the non-specific treatment for CML?
2. What is the targeted treatment for CML?
1. Hydroxyurea, cytarabine
2. Imatinib
What is the mechanism of action of Imatinib?
Imatinib binds to the ATP binding pocket of the Bcr-Abl fusion protein.
What is the common gene mutation for PV, ET, and PM?
JAK2
1. What happens to leukocyte and platelet counts in PV?
2. What happens to Epo in PV?
1. mild or no leukocytosis and/or thrombocytosis
2. Epo suppression
What are 4 non-clonal causes of elevated RBC mass?
1. Epo secretion from liver or kidney tumors or cysts or from leiomyomatas.
2. High altitude
3. Testosterone therapy
4. High affinity hemoglobinopathies
1. What is the function of mutant JAK2?
2. What is the normal substrate of JAK2?
Mutant JAK2 is constitutively active and does not require its normal substrate, Epo, for activation.
How does essential thrombocythemia lead to its clinical presentation?
Platelets derived from the abnormal megakaryocytes do not function properly, leading to bleeding and thrombosis.
Myelofibrosis can be caused by what other myeloproliferative neoplasms?
Polycythemia vera and essential thrombocythemia
What is the cure for myelofibrosis?
Allogeneic stem cell transplant
Tear drop cells are associated with what condition?
Myelofibrosis
80y female:
-abdominal pain
-portal venous thrombosis
-mild splenomegaly
-CBC: WBC 8, Hgb 14.9, Platelets 578,000
-Creatinine 1.4
1. What diagnostic tests would you order?
2. What is the most likely diagnosis?
1. Epo levels (if low=PV), JAK2 mutation, ESR/CRP (markers of inflammation)
2. PV
30y male
-acute appendicitis
-CBC: WBC 19,000 (mostly PMNs, bands), Hgb 12.3, Platelets 812,000
1. What is the most likely cause of the thrombocytosis?
1. Reactive inflammatory response to acute appendicitis
32y male
-CBC: WBC 7, Hgb 14.9, Platelet 970,000
1. What diagnostic tests would you order?
2. What is the most likely cause of the thrombocytosis?
1. ESR/CRP (markers of inflammation, JAK2 mutation
2. Essential thrombocythaemia
Are children more prone to getting ALL or AML?
ALL
What is the affect of acute leukemia on the bone marrow, organs, and system? How does this lead to the clinical presentation of acute leukemia?
BM failure causing:
-anemia, weakness, fatigue
-thrombocytopenia, bleeding
-neutropenia/leukopenia, infection
Organ/tissue infiltration causing:
-hepatosplenomegaly, lymphadenopathy
-gum hypertrophy
-bone pain
-CNS, skin, testis, any organ
Systemic symptoms
-Fever, sweats, weight loss
What is the diagnostic criteria for acute leukemia?
>20 (30%) blasts in the bone marrow
Which are myeloid markers in AML?
CD13
CD33
Which are pre B cell markers in ALL?
CD10 CALLA
CD19
CD20
Which myeloproliferative neoplasm has auer rods?
AML
1. What are FLT3 and NPM1 mutations associated with?
2. What is the prognostic effect of FLT3 and/or NPM1?
1. AML
2. FLT3 = poor prognosis
NPM1 = good prognosis
FLT3 + NPM1 = poor prognosis
1. What chromosome abberation is associated with APL?
2. What is the protein product of the chromosome abnormality and its function?
1. t(15;17)
2. PML-RARa fusion gene causes maturation arrest
Which form of AML is most easily cured?
APL
What are the two treatments for APL?
ATRA to promote differentiation
As<sub>2</sub>O<sub>3</sub> to kill of stem cell (hopefully)
What is the prognostic effect of the Ph chromosome on ALL?
bad
Which are monocytic markers in AML?
CD14
CD15
Which are megakaryocytic markers in AML?
CD41
CD62
Which are pre T cell markers in ALL?
CD2
CD3
CD5
CD7
1. What is the function of NPM1?
2. What is the effect of NPM1 mutations?
1. Nuclear/cytoplasmic shuttle
2. Mutant NPM1 cannot transport ribosomes out of the nucleus into the cytoplasm, decreasing protein synthesis
1. What is the prognostic indicator of MLL rearrangements in AML?
2. What phenotype of AML are MLL rearrangement associated with?
1. Intermediate risk
2. M4/M5 phenotypes
1. What is the function of FLT3?
2. What is the effect of FLT3 mutations?
1. Trans-membrane tyrosine kinase receptor.
2. Mutant FLT3 does not auto-inhibit and becomes a constitutive tyrosine kinase.
What is the standard remission induction therapy of newly diagnosed AML?
7+3 schedule
7 days Cytarabine infusion
Anthracycline on days 1-3
What is the first remission post-remission treatment of AML if <60y?
2-3 courses of high dose cytarabine
HSC transplantation in intermediate and high risk patients
1. What is a major clinical concern of APL?
2. What is the treatment?
1. Coagulopathy leads to bleeding
2. Platelet transfusion until DIC stops and cryoprecipitate to maintain fibrinogen
DIC is a hallmark of what leukemia?
APL
What are the molecular markers for plasma cells?
CD38
CD138
What is the most common indolent NHL?
Follicular lymphoma
What is the most common aggressive NHL?
Diffuse Large B-Cell Lymphoma
1. What is the most common B cell lymphoma in children?
2. What is the most common T cell lymphoma in children?
1. B Burkitt (Burkitt-like)
2. T Lymphoblastic
What are the two most common B cell lymphoma in adults?
1. Follicular BCL
2. DLBCL
What clinical presentation of lymphadenopathy is more likely to be benign?
Lymphadenopathy lasting <2w or >1y without a change in size has a low likelihood of being neoplastic.
What is the leukemia equivalent of B Cell lymphocytic lymphoma?
B CLL
What is the lymphoma equivalent of B CLL?
B Cell lymphocytic lymphoma
1. Which lymphoma is t(14;18) associated with?
2. What is the gene product?
3. What is the protein's effect?
1. Follicular Center Cell Lymphoma
2. Bcl-2
3. Antiapoptosis
1. Which chromosomal aberration is associated with Follicular Center Cell Lymphoma?
2. What is the gene product?
3. What is the protein's effect?
1. t(14;18)
2. Bcl-2
3. Antiapoptosis
1. Which lymphoma is t(8;14) associated with?
2. What is the gene product?
3. What is the protein's effect?
1. Burkitt Lymphoma
2. C-myc
3. Hyperproliferation
1. Which chromosomal aberration is associated with Burkitt Lymphoma?
2. What is the gene product?
3. What is the protein's effect?
1. t(8;14)
2. C-myc
3. Hyperproliferation
1. Which lymphoma is associated with t(11;14)?
2. What is the gene product?
3. What is the protein's effect?
1. Mantle Cell Lymphoma
2. Bcl-1/Cyclin-D1
3. Hyperproliferation
1. Which chromosomal aberration is associated with mantle cell lymphoma?
2. What is the gene product?
3. What is the protein's effect?
1. t(11;14)
2. Bcl-1/Cyclin-D1
3. Hyperproliferation
Which is more likely in young adults, HL or NHL?
HL has bimodal age distribution (20-29) and >65
Which is more likely in the elderly, HL or NHL?
NHL
Are males or females more prone to T lymphoblastic lymphoma?
Males
Are young or old people more prone to T lymphoblastic lymphoma?
Young people
Which virus is associated with Adult T Cell Leukemia/Lymphoma?
HTLV-1
Which lymphoma/leukemia is associated with HTLV-1?
Adult T Cell Leukemia/Lymphoma
Which form of HL is typically associated with young women?
Nodular sclerosing HL
Are women or men more prone to NSHL?
Women
1. Which cell line does GCSF increase?
2. What are GCSF's short-term and long-term mechanisms of action?
1. neutrophils
2. Release from BM (3-4h)
Increased production (3-5d)
Decreased apoptosis
What is Kostmann's syndrome?
Severe Congenital Neutropenia
Autosomal recessive disorder leading to lack of maturation of promyelocytes into neutrophils
Where are the majority of neutrophils?
In the bone marrow reserves
What happens to patients without any neutrophils?
Fulminant sepsis from regular ubiquitous organisms such as E. coli and Klebsiella spp. leading to death within 12h
Do patients with neutrophils in their bone marrow but not their circulation get infections?
No
How does CXCR4 mediate neutrophil maturation, life, and death?
CXCR4 is expressed as neutrophils mature in the bone marrow, internalized so they can enter the circulation, and externalized 1-3d later to re-enter the bone marrow for destruction.
What level of neutropenia poses a significant risk for infection?
ANC <300
What is a normal ANC for caucasians? For africans?
1500 for caucasians
1200 for africans
What should be considered in the case of neutropenia and anemia?
Possible malignancy
What compounds do neutrophil primary azurophilic granules contain?
Defensins
Myeloperoxidase
What compounds do neutrophil secondary specific granules contain?
NADPH-dependent neutrophil peroxidase
1. What is the defect in CGD?
2. What is its inheritance pattern?
3. How is it diagnosed?
1. Defective Phox gene creates inactive NADPH-dependent Neutrophil peroxidase
2. X-linked (2/3), recessive (1/3)
3. NBT test will not turn blue if CGD is present
In a population of more than 250,000,000 in the US, what is the approximate annual number of cancer deaths?
500,000
In a population of more than 250,000,000 in the US, what is the approximate annual incidence of cancer?
1,000,000
In a population of more than 250,000,000 in the US, what is the approximate annual number of lymphoma/leukemia deaths?
50,000
What is the marker phenotype of lymphocyte predominant HL?
CD20+
CD15-
CD30-
What is the marker phenotype of all classical HL?
CD20-
CD15+
CD30+
Which surface marker is found on follicular b cells?
CD20
1. What are B symptoms?
2. What causes B symptoms?
3. What is their effect on prognosis?
1. Fever, night sweats, weight loss >10%
2. IL-6, IL-1
3. Worse prognosis
1. What chromosomal aberration is associated with MALT?
2. What is the gene product?
3. What is the protein's effect?
1. t(11;18)
2. API2:MALT1 fusion protein
3. Antiapoptosis
1. What lymphoma is t(11;18) associated with?
2. What is the gene defect?
3. What is the protein's effect?
1. MALT
2. API2:MALT1 fusion protein
3. Antiapoptosis
What causes hemolysis in lymphoma?
Immune dysregulation
1. How is MALT associated with H. pylori infection cured?
2. What is the appropriate chemical therapy?
1. Treatment of the H. pylori
2. Proton pump inhibitor + clarithromycin + amoxicillin or metronidazole
Do plasma cells express CD20?
No, only follicular B cells express CD20
How are IL-6 and MM connected?
IL-6 is secreted by bone marrow stromal cells and supports the proliferation of plasma cells.
How are MIP-1 and MM connected?
Plasma cells secrete MIP-1, leading to increased osteoclast activity.
How are VEGF and MM connected?
Plasma cells secrete VEGF, leading to increased vascularization of their bone marrow microenvironment.
How does MM lead to humoral immune suppression and susceptibility to infection?
Multiple myeloma cells suppress the growth of normal plasma cells.
How are M-proteins or paraproteins characterized?
SPEP (serum protein electrotrophoresis) or UPEP (urine protein electrophoresis) reveals an M-spike.
S/UIEP (Serum/urine immunofixation electrophoresis) characterizes the type of immunoglobulin.
What are the S/S of MM and their 4 causes?
Anemia, thrombocytopenia, +/- neutropenia from BM replacement.
Bone pain and fractures from lytic lesions of bone.
Heart and kidney failure from monoclonal light chain deposition.
Hypogammaglobulinemia and increased risk of infection due to suppression of normal plasma cell growth.
What are 4 major plasma cell disorders characterized by a monoclonal spike?
Multiple Myeloma
Waldenstroms macroglobulinemia
Solitary Osseus Plasmacytoma
Amyloidosis
What are the CRAB diagnostic criteria for MM?
Hyper<b>c</b>alcemia
<b>R</b>enal Insufficiency
<b>A</b>nemia
Lytic <b>B</b>one lesions or osteoporosis
A patient with hypercalcemia and renal insufficiency may have what?
Multiple myeloma
What is the criteria for MGUS?
&lt;3g M spike<br>&lt;10% Plasma cells in BM
What is the criteria for smoldering MM?
<u>></u>3g M spike
<b>OR</b>
<u>></u>10% Plasma cells in BM
What is the criteria for active MM?
<u>></u>Positive M spike
<u>></u>10% Plasma cells in BM
<b>and</b>
Anemia, bone lesions, high calcium, or abnormal kidney function
Which two proteins are measured to determine the staging of MM?
Serum B<sub>2</sub>M
Serum albumin
What are the staging criteria for stage I MM?
Normal albumin (>3.5)
Low B<sub>2</sub>M (<3.5)
What are the staging criteria for stage II MM?
Albumin less than 3.5 + B<sub>2</sub>M less than 3.5
OR
Normal albumin with B<sub>2</sub>M 3.5-5.5
What are the staging criteria for stage III MM?
B<sub>2</sub>M greater than 5.5
1. What are Bence-Jones proteins?
2. Where are they found?
3. Why?
Bence-Jones proteins are light chain molecules from globulins usually found in the urine due to kidney damage leading to proteinuria.
Which molecules regulate osteoclast and osteoblast function?
RANKL activates osteoclasts
OPG (osteoprotegrin) is a decoy receptor for RANKL
DKK-1 inhibits osteoblasts
1. How are RANKL, OPG, and DKK-1 modified in MM?
2. What is the overall effect?
1. RANKL production is increased
OPG production is decreased
DKK-1 production is increased
2. The overall effect is a decrease in osteoblast differentiation and function and an increase in osteoclast differentiation and function.
Why are bisphosphonates given to those with MM?
Bisphosphonates inhibit DKK-1, the osteoblast inhibitor
What drugs inhibit DKK-1?
Bisphosphonates
What is the rate of conversion from MGUS to MM?
1-2% per year
What are the current chemotherapy treatments for MM?
Revlimid+dexamethasone
Velcade+dexamethasone
What is Revlimid and Dexamethasone used to treat?
MM
What is Velcade and Dexamethasone used to treat?
MM
How is MM progression or response measured?
The M-protein spike
What are the two most important factors in the viscosity of whole blood?
[RBCs]
[fibrinogen]
What are the most important factors in the viscosity of serum?
[proteins]
asymmetry of proteins
M.W. proteins
1. What is the cell of origin of Waldenstrom's Macroglobulinemia?
2. What is the general term for the tumor?
1. Lymphoplasmacyte
2. Lymphoplasmacytic lymphoma
What is the treatment for APL?
All-trans retinoic acid
What is the most common cause of APL?
Chromosomal translocation:
-Retinoic acid receptor alpha gene (RARa) on ch17
-Promyelocitic leukemia gene (PML) on ch15
What is the normal HCT range for men? For women?
HCT
Men: 39-51
Women: 37-47
What is the normal Hgb range for men? For women?
Hgb
Men: 13-17
Women: 12-16
What is the normal RBC range for men? For women?
RBC
Men: 4.5-6
Women: 4-5.3
How is MCV calculated? What is the normal range?
MCV = Hct/RBC*10
(80 - 100)
How is MCH calculated? What is the normal range?
MCH = Hgb/RBC*10
(28 - 33)
How is MCHC calculated? What is the normal range?
MCHC = Hgb/Hct*100
(31 - 35)
What is the normal reticulocyte range in percent and absolute count? How is the reticulocyte index calculated if shift cells are present?
.5-1.5%
30,000-70,000
If shift cells are present, reticulocyte index = reticulocyte count/2
What is the normal WBC count range?
WBC
5,000-10,000
What is the normal platelet count range?
Platelet
150,000-350,000
What is the normal plasma iron range?
Plasma Iron
70-150
What does TIBC measure? What is the normal range?
TIBC = Transferrin
250-350
What is the normal % iron saturation range?
15-45%
What is the normal ferritin range for men? For women?
Ferritin
Men: >100 ng/mL
Women: >20 ng/mL
Allergic reactions are mediated by which WBC?
Eosinophils
Hypersensitivity reactions are mediated by which WBC?
Basophils
What is the prognosis of untreated acute leukemia? How differentiated are the cells in acute leukemia?
Acute leukemia
Death in &lt;6mo if not treated
Blast cells with little or no differentiation
What is the prognosis of untreated chronic leukemia? How differentiated are the cells in chronic leukemia?
Chronic leukemia
Death in >1y if not treated
Mature cells more differentiated than blast cells
A leukemia composed of precursors of one or more of the following cell lines is referred to as which type of leukemia:
Erythroid
Neutrophil
Monocyte
Eosinophil
Basophil
Megakaryocyte
Myeloid leukemia
A leukemia composed of precursors of one or more of the following cell lines is referred to as which type of leukemia:
T Cell
B Cell
Lymphoid leukemia
Describe the leukemic phase of lymphoma
When the lymphocytic cells of a lymphoma disseminate and involve the bone marrow and peripheral blood, resembling an acute or a chronic leukemia, depending on the stage of differentiation of the lymphocyte
Describe a plasma cell myeloma
A clonal proliferative disorder of plasma cells which usually involves the bone marrow and presents with lytic bone lesions, but can sometimes present as plasma cell leukemia or progress into a leukemic phase
What does this image of bone marrow show?
What does this image of bone marrow show?
Aplastic anemia
What is the ddx of aplastic anemia including hereditary causes and acquired causes (direct toxicity, indirect toxicity, and unknown)?
<b>Hereditary</b>
-Fanconi's anemia
<b>Acquired</b>
<u>Direct Toxicity</u>
-Radiation
-Chemotherapeutic drugs
-Toxins (benzene)
<u>Indirect toxicity</u>
-Idiopathic
-Drugs (chloramphenicol)
-Infections (TB, HIV, EBV, CMV, HBV)
-Pregnancy
<u>Unknown</u>
-Paroxysmal Nocturnal Hemoglobinuria
What mechanism is probably required for indirect toxicity causing aplastic anemia?
Immune mediation
What are the two treatments for Aplastic anemia?
Best: Bone marrow transplantation with matched sibling HLA-type
If no matched sibling is available, then:
Antithymocyte globulin + high dose steroids x2 weeks with taper
Cyclosporin A x1-2 years
This destroys T cells, then WBC comes back, then RBC comes back
If unsuccessful with pharmacologic intervention, use matched unrelated donor
Describe feedback regulation of heme synthesis in nonerythroid tissue. Which isoform is present here?
Heme inhibits ALA-synthase 1 (ALA-S1) activity and gene expression, required for the first step in heme synthesis, and mitochondrial translocation
What is the first step of heme synthesis? What enzyme catalyzes the reaction? What co-factor is required? How is this step regulated? Where does this reaction occur?
glycine + succinyl CoA --> d-aminolevulinicacid + CoASH
+ CO2
ALA synthase
Vitamin B6 (Pyridoxal phosphate)
Inhibited by heme
Mitochondria
What other enzymes require heme?
Cyt P450
Catalase
Mitochondrial cytochromes
What is the metabolic capacity of RBCs?
Glycolysis
Pentose Phosphate Pathway
How many moles of glycine and succinyl CoA are required for one molecule of heme?
8
What is the final enzymatic step of heme synthesis? What enzyme catalyzes this reaction? Where does this reaction occur?
Protoporphyrin IX + Fe<sup>2+</sup> --> Heme + 2H<sup>+</sup>
Ferrochetalase
Mitochondria
Describe feedback regulation of heme synthesis in erythroid tissue. Which isoform is involved here?
The 5' UTR of the ALA-S2 mRNA contains IREs, which are tightly bound to IRP in the absence of iron, blocking translation.
When iron is present, the IRP binds to iron and cannot bind to the 5' UTR of ALA-S2, therefore translation occurs
How is expression of the transferrin receptor regulated?
In the absence of iron, IRP bind to IRE in the 3' UTR of transferring mRNA, increasing the stability of the mRNA.
In the presence of iron, Fe binds to the IRP, exposing the IRE in the 3' UTR leading to unstable mRNA which is degraded.
What is the cause of Congetinal Erythropoietic Porphyria?
What compound builds up?
What is the treatment?
Congenital Erythropoietic Porphyria
Cause: Deficiency of Uroporphyrinogen III cosynthase activity
Accumulation: Uroporphyrinogen I/Coporphyrinogen I
Tx: sun avoidance, skin protection, marrow suppression, marrow transplantation, possible gene therapy, possible I.V. hematin
What is the cause of Acute Intermittent Porphyria?
What compound accumulates?
What is the treatment?
Which drugs does this condition contraindicate?
Acute Intermittent Porphyria
Cause: Deficiency of porphobiliogen deaminase
Accumulation: d-ALA, porphobilinogen
Tx: hematin
Contraindicates: stimulators of ALA synthase and ALA dehydratase
Which form of Hb is taught?
deoxy
Which form of Hb is relaxed?
oxy
Which has a greater P<sub>50</sub>, Hemoglobin or myoglobin?
Hemoglobin
1. What is the hill coefficient of myoglobin?
2. What is the hill coefficient of oxygen?
3. What does this mean?
1. Myoglobin = 1.0
2. Hemoglobin = 2.8
3. Hemoglobin is cooperative
What is P<sub>50</sub>?
The concentration at which 50% of the units are occupied
What is the normal adult isoform of hemoglobin? what is its relative abundance?
HgbA<sub>1</sub>: alpha<sub>2</sub>beta<sub>2</sub>
~92%
What are the subunits of HbA<sub>2</sub>? What is its relative abundance?
HgbA<sub>2</sub>: alpha<sub>2</sub>delta<sub>2</sub>
~2.5%
What is the isoform of hemoglobin caused by non-enzymatic glycosylation? What is its normal relative abundance?
HgbA<sub>1c</sub>: alpha<sub>2</sub>beta<sub>2</sub><sup>a-N-glucose</sup>
~5%
What is the fetal isoform of hemoglobin? What is its normal relative abundance?
HgbF: alpha<sub>2</sub>gamma<sub>2</sub>
&lt;1%
What sort of region in the globin chain does heme reside? Why?
Hydrophobic pocket
Protects Fe2+ from oxidizing to Fe3+ due to water
How does O<sub>2</sub> binding to hemo affect the tertiary structure of hemoglobin?
When O<sub>2</sub> is not bound, the iron is out of the heme plane. Once O<sub>2</sub> binds, the iron moves into the heme plane and pulls the proximal His ligand along with it, causing a minimal change (.29A) which increases overall O<sub>2</sub> binding cooperatively.
What two chemical reactions are important for the Bohr Effect? Which way does it shift the Oxygen dissociation curve? What are the two outcomes?
Hb(O<sub>2</sub>)<sub>4</sub> + xH<sup>+</sup> --> Hb<sub>4</sub> + 4O<sub>2</sub> [x=1.8-2.8]
CO<sub>2</sub> + H<sub>2</sub>O <--> H<sub>2</sub>CO<sub>3</sub> <--> HCO<sub>3</sub><sup>-</sup> + H<sup>+</sup>
Right Shift
1. Easier release of O<sub>2</sub> in tissues where CO<sub>2</sub> creates an acidic environment
2. Easier venting of CO<sub>2</sub> in the lungs where O<sub>2</sub> tension pushes the H<sup>+</sup> to form CO<sub>2</sub> and H<sub>2</sub>O
What is the biochemical mechanism of the Bohr effect?
Protonation at low pH stabilizes the deoxy taught form of hemoglobin
What is the effect of DPG (BPG, 2,3-diphosphoglycerate) on the oxygen dissociation curve of hemoglobin? Where does DPG bind to the hemoglobin molecule?
DPG binds between the two b-chains on + charged amino acids
What chemical reaction is important for the effect of CO<sub>2</sub> binding to hemoglobin? Where does CO<sub>2</sub> bind to hemoglobin? What is the result of that binding?
CO<sub>2</sub> + H<sub>2</sub>N-Hemoglobin <--> <sup>-</sup>O(C=O)NH-Hemoglobin
1. CO<sub>2</sub> binds to deoxy Hb more than oxy Hb
2. CO<sub>2</sub> binds to a-amino groups of b-chains more than a-chains
3. Carbamino group forms a salt bridge with + charged groups in deoxy form to stabilize deoxy form
How does temperature affect the oxygen dissociation curve?
Higher temp shifts the curve to the right, lower temp shifts the curve to the left
What are the genetic mechanisms of hemoglobinopathies?
Single point mutation (sick cell HbS)
Double Amino Acid Substitution (HbC Harlem)
Variants with deletions (Hb Gun Hill)
Chain Termination mutants (Hb Constant Spring)
Frame Shift Mutations (Hb Wayne)
Fusion Chain (Hb Lepore)
Insertion (Hb Grady)
What is the mutation for sickle cell anemia? How does the HbS contribute to sickling?
AA#6 glu --> val (hydrophilic to hydrophobic change)
Hbs has lower O2 affinity, releases O2 easier to account for anemia
deoxyHbS aggregates, blocking capillaries and causing stasis
Stasis --> acidosis --> oxygen curve shifts right --> O<sub>2</sub> release --> more sickling
Which two hemoglobinopathies lead to denatured hemoglobin inclusions? What are these called?
Hb Hammersmith (severe)
Hb Koln (mild)
Heinz Bodies
What are the possible causes of methemoglobinemia?
Acquired/Toxic: Drug-induced oxidation of HbA
Hereditary HbM: distal or proximal His --> Tyr mutation
Hereditary NADH Cytochrome B5 Reductase deficiency
How much ATP is generated per mole of glucose? What is ATP's purpose in the RBC?
1 mole glucose --> 2 mole ATP
ATP powers the Na<sup>+</sup>/K<sup>+</sup>ATPase and the Ca<sup>2+</sup>ATPase to keep intracellular sodium low, potassium high and calcium low
How do SS RBCs become dehydrated and rigid?
SS RBCs have reduced Ca<sup>2+</sup>ATPase activity which causes increased rigidity and increases the influx of calcium which causes the Gardos effect and opening of K<sup>+</sup> channels leading to dehydration.
Heterozygous patients with a partial NADH Cytochrome B5 Reductase deficiency should not be given what drugs? Why?
Oxidant drugs (primaquine, chloroquine)
They will develop acute methemoglobinemia due to hemoglobin oxidation and reduced reduction.
How does a pyruvate kinase deficiency lead to hemolytic anemia?
Pyruvate kinase deficient patients have ~5-25% of normal PK activity, thus their rate of glycolysis is decreased and less ATP is produced.
What are the first and second most common causes of hemolytic anemia?
1. G-6-P Dehydrogenase deficiency
2. Pyruvate Kinase deficiency
Where are phosphatidyl choline, sphingomyelin, phosphatidyl ethanolamine, and phosphatidyl serine found in the RBC membrane? What happens if PS is in the wrong leaflet?
PC and SM are in the outer leaflet
PE and PS are in the inner leaflet
If PS is in the outer leaflet, as can occur in sick cell disease, increased coagulation can occur.
What is the cause of hereditary spherocytosis? What is the clinical result?
Autosomal dominant spectrin deficiency leads to mild anemia.
What is the cause of hereditary elliptocytosis? What is the clinical result?
Autosomal dominant failure of spectrin molecules to form tetramers leads to mild anemia
What is the cause of acanthocytosis? What patients are at risk??
Autosomal recessive increase in cholesterol and sphingomyelin increases RBC rigidity and leads to hemolytic anemia, more often in patients with liver disease especially due to chronic alcoholism.
What conditions increase the propensity of HbS to polymerize and cause sickling?
Increased Temp (fever)
Low pH (acidosis)
Dehydration
Hypoxia (stasis, high elevation)
What are the treatment options for Sickle Cell Disease (SCD)?
Lower fever
Correct acidosis
Increase fluid intake
Hydroxyurea increases fetal HbF (alpha2gamma2), known teratogen
Prevent adhesion of SS RBC to endothelium
Chronic hemolytic anemia, vaso-occlusive crises, frequent infections, leukocytosis, pulmonary hypertension, and asthma are clinical manifestations of what disease?
Sickle Cell Disease
What are the 4 major mechanisms of anemia due to chronic disease and/or inflammation?
1. Activation of macrophages and spleen
2. Faulty iron re-utilization
3. Blunted EPO response
4. Cytokine inhibition of erythropoiesis
How do proinflammatory cytokines lead to RBC lifespan being decreased by 10-20d?
Proinflammatory cytokines (TNF-a) upregulate macrophage and splenic function and cause RBC membrane changes, leading to erythrophagocytosis
How does chronic disease and/or inflammation lead to faulty iron-reutilization?
Lactoferrin is released from PMNs which selectively transports iron to macrophages.
The liver synthesizes hepcidin which reduces intestinal absorption and transferring production while increasing macrophage retention of iron as ferritin, decreasing serum iron levels.
How does chronic disease and/or inflammation lead to a blunted EPO response?
IL-1 and TNF-a directly inhibit EPO mRNA via ROS that damage EPO producing cells
Pancytopenia with decreased reticulocyte count, increased MCV RDW LDH and indirect Bilirubin is what form of anemia? What type of anemia is this?
Megaloblastic anemia
Ineffective hematopoiesis
How is B12 deficiency a self-impairing cycle?
Vitamin B12 is especially required in cells with a high turnover, such as gut epithelial cells. Atrophy of gut cells due to a lack of B12 leads to decreased absorption of B12
What is the cause of pernicious anemia?
Autoimmune antibody to gastric H+/K+ ATPase --> gastric atrophy
Ab to intrinsic factor are specific to what disease? How specific?
Pernicious anemia
~100% specific
How many base pairs are in the human genome?
3 billion
How many genes are in the human genome?
25,000-35,000
By how much do individuals differ from one another genetically?
1 base per 1000
What is the difference between an early myeloblast and a PMN in terms of phenotypic markers?
Early myeloblast: CD33+, CD34+
PMN: CD33+, CD34-
What is the most sensitive method of genetic detection?
PCR (1/1,000,000)
1. How is the immune system dysregulated in Hodgkin Disease?
2. What sort of infections are individuals prone to?
1. T cell function appears to be abnormal
2. Patients have increased susceptibility to TB, L. monocytogenes, Candida, Herpes simplex and to opportunistic infections by P. carinii, vaccinia, and herpes zoster
1. How does HD spread typically?
2. How does HD spread in HIV+ patients?
1. HD typically spreads in a linear nodal fashion (mediastinal mass-->axillary/cervical nodes)
2. Skipped nodes are more common (retroperitoneal-->supraclavicular)
Describe liver and splenic involvement in HD
HD can involve the spleen, and often will also involve the liver
HD rarely involves the liver only
What is the differential diagnosis of a mediastinal mass?
4Ts
Thyroid (substernal goiter)
Teratoma
Thymoma
Terrible lymphomas
-T Cell lymphoma/leukemia
-B cell lymphoma
-Hodgkin lymphoma
1. What is the role of APOBEC3?
2. How does HIV block APOBEC3?
1. APOBEC3 deaminates C to U in retroviral cDNA, leading to fatal mutations
2. HIV Vif inhibits APOBEC3
1. What is the role of HIV Vif?
Vif inhibits APOBEC3 from deaminating C to U in retroviral cDNA
1. What is the role of TRIM5a?
TRIM5a restricts HIV infection in other primates and prevents capsid shedding and release of viral RNA
1. What is the role of Tetherin/CD317?
2. How does HIV block Tetherin?
1. Tetherin prevents HIV virion release from the cell membrane
2. HIV Vpu inhibits tetherin
What is the role of HIV Vpu?
Vpu inhibits tetherin/CD317 from preventing the release of HIV virion particles
What is the role of HIV Vpr?
HIV Vpr blocks IL-12 production by macrophages
What is the role of Nef?
Nef blocks CD4 expression and inhibits apoptosis in infected cells
What are the two ways in which HIV depletes CD4+ T cells?
1. Direct cytotoxicity from virion production
2. Syncytia formation from gp120 binding to other T cell CD4 and causing membrane fusion
During the acute HIV infection, which test is the most likely to confirm a diagnosis of HIV?
HIV viral load will detect the rapid rise in viral load which follows from HIV innoculation and acute HIV syndrome, before the immune system reduces the viral load.
A patient with fevers, chills, and a maculopapular rash x3w saying "this is the worst cold I've ever had" has what condition?
Acute retroviral syndrome from HIV infection
When will HIV viral RNA become detectable in a patient?
2-4w (5-14days)
When will viral load HIV RNA become positive in a patient?
2-4w (5-14d)
When will anti-HIV ab become positive in a patient using EIA?
+/- @ 4-6w
+ @ 2mo
When will anti-HIV ab become positive in a patient using western blotting?
+/- @ 2mo
+(3) @ 3mo
+(8) @ 6mo
What are the clinical signs of chronic, symptomatic HIV in men? In women?
Men: Wasting (loss of >15% bw), Thrush, Fever>2w, Diarrhea >1mo, ITP, Oral hairy leukoplakia, Hepes Zoster
Women: Same as men plus Recurrent Vulvovaginal Candidiasis, Cervical dysplasia, CA in situ of cervix, PID
Why is HIV associated with cervical dysplasia and CA in situ of the cervix?
There is a high rate of HPV + HIV coinfection
At what CD4 levels can herpes zoster occur in HIV?
Any
Which virus is oral hairy leukoplakia associated with?
EBV
Which test is most likely to confirm a diganosis during a chronic HIV infection?
Western blot for anti-HIV ab
What is the lethal disease and causative organism in AIDS?
PCP (pneumocystis pnuemonia) caused by pneumocystis jiroveci
What is the clinical defintion of AIDS?
Any of the following
CD4 <200
CD4 <15%
Opportunistic infection or disease
Is invasive cervical cancer a sign of chronic HIV or AIDS?
AIDS
When is a patient at risk for PCP?
CD4<200
What is the treatment of PCP?
TMP/SMX x21d + Prednisone
When is a patient at risk for CMV retinitis?
CD4 <50
1. When is an HIV patient at risk for Kaposi Sarcoma?
2. What is the causative agent of kaposi sarcoma?
1. Any CD4 level
2. HSV 8
When is a patient at risk for Cryptococcus meningitis?
CD4<100
How is cryptococcus meningitis diagnosed?
India Ink stain of LP, confirmatory CSF and serum crypto antigen titers
When is a patient at risk for CNS toxoplasmosis?
CD4<100
How is CNS toxoplasmosis diagnosed?
multiple ring enhancing lesions
How is cryptococcus meningitis treated?
Amphotericin and 5-flucytosine followed by diflucan
How is toxoplasmosis treated?
sulfadiazine, pyremethamine, leukovorin
Lymphoma of the brain causing a CNS mass lesion is associated with what virus?
EBV
Who should be tested for HIV under CDC testing recommendations?
All peoples aged 13-64
Anyone:
-tested or treated for an STD
-initiating TB tx
-pregnant women
-women with undocumented HIV status at L&D
Is written informed consent for HIV testing required?
no
Who should be started on ART?
Strong recommendation: AIDS or CD4 <300, pregnant, chronic HepB, or HIV nephropathy
Moderate-strong recommendation: CD4 350-500
50% optional, 50% in favor: CD4 >500
1. What is the mechanism of action of Emtricitabine?
2. What are its side effects?
1. Nucleoside reverse transcriptase inhibitor for the treatment of HIV
2. None
1. What is the mechanism of action of Tenofovir?
2. What are its side effects?
1. Nucleoside reverse transcriptase inhibitor for the treatment of HIV
2. Decreased mineral bone density, rare fanconi syndrome
1. Which two nucleoside reverse transcriptase inhibitors are common prescribed during HAART?
2. What are their side effects?
Emtricitabine - None
Tenofovir - decreased mineral bone density, rare fanconi syndrome
1. What is the mechanism of action of Atazanavir?
2. What are its side effects?
1. Protease inhibitor for the treatment of HIV
2. increased indirect bilirubin, prolonged PR, CYP3A4 inhibition
1. What is the mechanism of action of Darunavir?
2. What are its side effects?
1. Protease inhibitor for the treatment of HIV
2. Rash, CYP3A4 inhibition
1. Which two protease inhibitors are commonly prescribed during HAART?
2. What are their side effects?
Atazanavir - increased indirect bilirubin, prolonged PR, CYP3A4 inhibition
Darunavir - rash, CYP3A4 inhibition
1. What is the mechanism of action of Efavirenz?
2. What are the commonly associated side effects of Efavirenz?
1. Non-nucleoside reverse transcriptase inhibitor for the treatment of HIV
2. CNS side effects, rash, CYP3A4 induction
1. What is the common side effect of nucleoside reverse transcriptase inhibitors?
2. What are two commonly prescribed NRTIs?
CYP3A4 inhibition
Atazanavir
Darunavir
Which drugs inhibit CYP3A4?
Atazanavir
Darunavir
Nucleoside reverse transcriptase inhibitors
Which drug induces CYP3A4?
Efavirenz
non-nucleoside reverse transcriptase inhibitor
1. What is the mechanism of action of Raltegravir?
2. What are its side effects?
1. Integrase inhibitor for the treatment of HIV
2. none common or severe, headache, nausea, and diarrhea can occur
What are the antiretroviral guidelines?
2 nucleoside RT inhibitors (emtricitabine + tenofovir)
Plus one of the following:
-protease inhibitor(s) atazanavir or darunavir
-non-nucleoside RT inhibitor efavirenz
-integrase inhibitor raltegravir
1. Which drug inhibits integrase?
2. What are its side effects?
Raltegravir - none common or severe, headache, nausea, diarrhea may occur
What is the current recommended regimen for HIV+ pregnant women?
Lopinavir/ritonavir + AZT/3TC(lamivudine)
1. What is the mechanism of action of Chloroquine, quinine, quinidine, and mefloquine?
2. Are they blood schizonticides or hepatic hypnozoiticides?
1. Drug is concentrated in infected erythrocytes and raises the pH in the food vacuole of the parasite, preventing heme polymerization to hemozoin; heme accumulates and kills the parasite.
2. Blood schizonticides
What is the mechanism of action of primaquine?
2. Is it a blood schizonticide or a hepatic hypnozoiticide?
1. Redox cycle generates oxidants, interferes with electron transport chain in mitochondria.
2. Hepatic hypnozoiticide
Which antimalarial drug is safe for pregnant women?
Chloroquine
Which antimalarials should not be combined?
Mefloquine with any other quinoline (quinine, quinidine, chloroquine)
Which antimalarial drug contributes to oxidative damage of RBCs?
Primaquine
1. What is the mechanism of action of Pyrimethamine?
2. What is the mechanism of action of sulfadoxine?
3. What are they used to treat?
1. Pyrimethamine inhibits DHFR
2. Sulfadoxine inhibits dihydropteroate synthase
3. They are used to treat Chloroquine-resistant P. falciparum
1. What is the mechanism of action of Proguanil?
2. What is the mechanism of action of Atovaquone?
3. What are they used to treat?
1. Proguanil inhibits DHFR
2. Atovaquone binds to Cyt bc1 and inhibits electron transport, collapsing the mitochondrial membrane potential.
3. Proguanil/Atovaquone (malarone) are used to treat chloroquine and MDR P. falciparum
Describe the synergy of Proguanil and Atovaquone
Proguanil inhibits DHFR, but also enhances atovaquone. Atovaquone binds to Cyt bc1 and collapses the mitochondrial membrane potential.
What is the WHO recommendation for the treatment of falciparum malaria?
Artemisin-based combination therapy for 3 days (Artemether+lumefantrine or Artesunate+something else)
What is the WHO recommendation for the treatment of severe malaria?
Artesunate (quinine or quinidine in the US)
What is the diagnostic test for malaria?
Thick and thin smears of blood
Which forms of malaria form hypnozoites?
P. vivax
P. ovale
1. What is the treatment recommendation for P. vivax or P. ovale?
2. What are the contraindications for this treatment?
1. Chloroquine x3d + Primaquine x14d
2. Primaquine is contraindicated in those with G6PD deficiency
Coma, metabolic acidosis, severe anemia, hypoglycemia, acute renal failure, and pulmonary edema due to a parasitic infections are signs of infection by what organism and what disesae?
Severe malaria due to P. falciparum
Which forms of malaria cause a tertian fever?
P. vivax
P. ovale
P. falciparum
Which forms of malaria cause a quartan fever?
P. malariae
What causes the fevers in malaria?
TNF-a release due to macrophages being stimulated by gycosyl phosphatidylinositol moieties as the schizonts rupture in the blood
What are 3 causes of thrombocytosis?
1. Fe deficiency
2. Chronic disease
3. Essential thrombocytopenia
4. Cancer
In what conditions is the RDW not increased?
Thalassemia minor
Anemia of chronic disease
Aplastic anemia
Which conditions cause a microcytic anemia?
Fe deficiency
Thalassemia
Anemia of chronic disease (20%)
Sideroblastic anemia
Which conditions cause a macrocytic anemia?
Sideroblastic anemia
Pernicious anemia
Folate deficiency
Liver disease
Which conditions cause a normocytic anemia?
Aplastic anemia
Blood loss
Anemia of chronic disease (80%)
For which patients is HAART strongly recommended?
AIDS
CD4 <350
Pregnancy
Chronic Hepatitis B needing tx
HIV nephropathy
For which patients is there a 55% strong and 45% moderate recommendation for HAART?
CD4 350-500
For which patients is there a 50% favorable and 50% optional view of HAART?
CD4 >500
What is the mechanism of action of enfuvirtide?
HIV fusion inhibitor
What is the mechanism of action of T-20?
HIV fusion inhibitor
What is the mechanism of action of maraviroc?
CCR5 receptor antagonist
What are the 3 possible regimens and their side effects for HAART?
2xNRTIs: Emtricitabine (none) + Tenofovir (decreased mineral bone density)
and 1 of the following:
NNRTIs: Efavirenz (CNS, rash, hypersensitivity)
OR
Integrase Inh: Raltegravir (none common or severe, HA, N, D)
OR
Protease inhibitors: Darunavir/r (Rash) or Atazanavir/r (PR elongation, increased indirect bilirubin)
What is the HAART for pregnant women?
LPV/r + AZT/3TC
Describe the important steps in heme synthesis
<b>Mitochondria</b>
Sucincyl Coa + Glycine -[d-ALA synthase]-> d-ALA
<b>Cytoplasm</b>
2x d-ALA -[PBG synthase]-> PBG
4x PBG -[PBG deaminase]-> HMB
HMB -[Uroporphyrinogen III Cosynthase]-> Uroporphyrinogen III
Uroporphyrinogen III --> Coproporphyrinogen III
<b>Mitochondria</b>
Coporporphyrinogen III --> Protoporhyrin IX
Protoporphyrin IX -[Ferrochetalase]-> Heme
1. What is the deficient enzyme of AIP?
2. What step of heme synthesis is affected?
3. What molecules build up?
4. What is the treatment?
1. PBG deaminase
2. 4x PBG -[PBG deaminase]-> HMB
3. PBG and d-ALA
4. IV hematin
1. Deficiency of PBG deaminase is associated with what condition?
2. What molecules build up?
3. What should be avoided?
4. What is the treatment?
1. AIP
2. PBG and d-ALA
3. drugs, steroids, dietary changes, inducers of d-ALA synthase
4. IV hematin
1. What is the deficient enzyme of CEP?
2. What step of heme synthesis is affected?
3. What molecules build up?
4. What is the treatment?
1. Uroporphyrinogen III cosynthase
2. HMB -[Uroporphyrinogen III cosynthase]-> Uroporphyrinogen III
3. Uroporphyrinogen I and Coproporphyrinogen I
4. Transferrin suppresses defective erythropoiesis, IV hematin
1. Deficiency of Uroporphyrinogen III cosynthase is associated with what condition?
2. What step of heme synthesis is affected?
3. What molecules build up?
4. What is the treatment?
1. CEP
2. HMB -[Uroporphyrinogen III cosynthase]-> Uroporphyrinogen III
3. Uroporphyrinogen I and Coporporphyrinogen I
4. Transferrin suppresses defective erythropoiesis, IV hematin
What does a right shifted O2 saturation curve reflect?
Decreased affinity
What is the effect of increased temperature on the O2 saturation curve?
Right shift, decreased affinity
What is the effect of decreased pH (increased [H<sup>+</sup>]) on the O2 saturation curve?
Right shift, decreased affinity
What is the effect of no DPG on the O2 saturation curve?
Left shift, increased affinity (fetal Hb)
Which HAART drugs induce CYP3A4?
nNRTIs
Efavirenz
What are the symptoms of cinchonism?
flushing
sweaty
tinnitus
dizziness
nausea
diarrhea
What are the causes of microangiopathic hemolytic anemia?
DIC
TTP
Vasculitis
Mechanical heart valves
What is the cause of IIa VWD?
Absence of HMW vWF mutimers
Which leukemia is often the cause of ITP?
CLL
When are spur cells often visible?
Cases of liver failure
1. What are platelet concentrates used to treat?
2. What are they not used to treat?
1. Thrombocytopenia
2. ITP
What are FFP used to treat?
Factor deficiencies
When are target cells visible?
Liver disease
thalassemia
When are burr cells visible?
Kidney disesae
1. How many copies of Hb-a are there in a-thal minima?
2. What is the other name for this disease?
1. 3
2. Thal-2
1. How many copies of Hb-a are there in a-thal minor?
2. What is the other name for this disease?
1. 2
2. Thal-1
1. What is HbH?
2. What is the effect of HbH on the O2 dissociation curve?
3. In what disease is HbH seen?
1. HbB<sub>4</sub>
2. Major left shift (increased affinity)
3. HbH disease/a-Thal intermedia
What is the mutation responsible for sickle cell anemia?
AA6 of HbB: Glu-->Val
What are the two reasons why NO is reduced in sickle cell anemia?
1. RBCs are lysed and produce arginase, which degrades L-arginine. NO is produced from L-arginine.
2. Free heme binds to NO in the bloodstream
What is contained within platelet primary granules?
Sticky stuff
What is contained within platelet dense granules?
ATP, ADP, serotonin, calcium
1. What platelet receptor does vWF bind to?
2. A lack of this receptor is known as what disease?
1. GpIb/IX
2. Bernard-Soulier Syndrome
1. What platelet receptor cross-links platelets with fibrin?
2. A lack of this receptor is known as what disease?
1. GpIIb/IIIa
2. Glanzmann's thrombasthenia
What is the cause of type IIb vWD?
Lack of HMW vWF multimers and increased affinity for platelet gpIb
What is the cause of type IIn?
vWF variants have markedly decreased affinity for VIII, VIII is degraded.
In which form of vWD will platelets be low?
vWD IIb
In which form of vWD will there be no FVIII?
vWD IIn
A lack of ADAMTS13 leads to what disorder?
TTP
What is gray platelet syndrome?
Deficiency of alpha granules leads to lifelong mucocutaneous bleeding
Low LDH levels in a sick cell patient often indicate what sort of disease progression?
Pain crises
Which chromosome is Hb-b on?
Ch11
What are the four mechanisms by which anemia of chronic disease occurs?
1. Shortened RBC survival due to macrophage activation
2. Lactoferrin moves iron to macrophages and hepcidin causes retention in macrophages and decreased GI absorption of iron
3. Blunted EPO response by IL-1 and TNF-a
4. IFN-g inhibits CFU-E growth, IL-1 inhibits CFU-E growth, TNF-a decreases stem cell colony formation
1. What is a normal PT time?
2. What is the criteria for abnormal PT?
Normal: 12 seconds
Abnormal: >1.3
1. What is a normal aPTT?
2. What is the criteria for abnormal aPTT?
1. Normal: 35 seconds
2. Abnormal: >1.3
1. What is a normal TT?
2. What is the criteria for abnormal TT?
1. Normal: 30 seconds
2. Abnormal: >1.3
1. What does a deficiency in ADAMTS13 cause?
2. What is the effect on platelets?
TTP
Increased aggregation of platelets due to increased HMW-vWF
Which will lead to a low bleeding time, TTP or ITP?
ITP will have a normal bleeding time even though there are few platelets
TTP will have a prolonged bleeding time
Which factors required for primary hemostasis will kidney failure lead to a deficiency of?
vWF
Fibrinogen
What will a deficiency of FXIII cause?
Delayed bleeding since XIIIa is responsible for cross-linking fibrin
What will a deficiency of FXII cause?
Thrombotic disorder
What will a deficiency of FXI cause?
Bleeding related to trauma, more common in the Jewish population
Factor V Leiden will lead to thrombosis in which vascular system?
venous
How is antiphospholipid characterized?
Why is it paradoxical?
Lupus anticoagulant is an antibody inhibitor directed at membrane phospholipids which prolongs PTT and PT but leads to thrombosis.
What differentiated liver disease from vitamin k deficiency?
FV is low in liver disease, not vitamin K deficiency
1. What is the cause of Tranmissable Spongiform Encephalopathy?
2. What is the name of the disease in humans?
1. Prions
2. Creutzfeldt-Jacob Disease
What are the three forms of Creutzfeldt-Jacob Disease?
Spontaenous
Inherited (Prnp mutation)
Acquired (vCJD, iatrogenic, transfusion, eating infected brain tissue)
1. What does the Prnp gene code for?
2. What is the normal characteristics of the protein product and its name?
3. What is the abnormal characteristics of the disease protein product and its name?
1. PrP
2. PrP<sup>C</sup>, many a-helices
3. PrP<sup>SC</sup>, many b-sheets
What is the structure of CMV?
Enveloped, icosahedral, dsDNA genome herpesvirus
How does CMV evade the immune system?
Suppresses MHC I/II expression
Produces proteins which block NK attack
IL-10 downregulates Th1 response
1. Describe the structure of West Nile Virus
2. What are the common, uncommon, and rare presentations of West Nile Virus?
1. ss(+)RNA
2. 80% asymptomatic, 20% West Nile Fever, 1% neuroinvasive
1. Describe the structure of West Nile Virus
2. How is West Nile Virus diagnosed?
1. ss(+)RNA
2. WNV IgM/IgG antibodies in blood or CSF
At what CD4 level does a person have AIDS?
<200
At what CD4 level does a person have symptomatic HIV disease?
<500
at what CD4 level does a person have PCP?
<200
At what CD4 level does a person get cryptococcus meningitis?
<100
At what CD4 level does a perso get toxoplasmosis or candidal esophagitis (disseminated cadidiasis)?
<100
At what CD4 level does a person get CMV retinitis?
<50
What are the most common pathogens which cause nosocomial pneumonia?
S. aureus
P. aeruginosa
What are the 4 most common Hospital-Acquired infections in order of increasing prevalence?
1. UTI
2. Pneumonia
3. Central line associated blood infection
4. Surgical Site infection
What are the most common pathogens which cause Central Line Associated Blood infections?
Coagulase(-) Staph (epidermidis), S. aureus, Enterococci, Candida
Which operations have the highest risk of Surgical Site infections?
Colon, small bowel, gastric operations
What are the most common pathogens which cause surgical site infections?
S. aureus, Coagulase(-) staph, Enterococci, E. coli
1. What is SIRS?
2. What is the clinical definiciont of SIRS?
1. Systemic Inflammatory Response Syndrome
2. 2+ of the following:
-T >38C or &lt;36c>90bpm
-Resp >20
-WBC >12,000 or &lt;4000>10% bands
1. What is the clinical definition of Sepsis?
SIRS + Infection
What is the clinical definition of severe sepsis?
Sepsis (SIRS + infection)
Organ dysfunction
Hypoperfusion or hypotension (systolic &lt; 90mmHg)
May include lactic acidosis, oliguria, AMS, or aute lung injury
What is the clinical definition of septic shock?
Sepsis (SIRS + infection)
Hypotension despite fluid rescucitation
1. What is the primary site of infection in sepsis?
2. Where does the infection often spread?
1. Lung
2. Spread to blood and abdomen
1. Which TLR recongizes LPS?
2. Which signalling molecules are released?
1. TLR-4
2. TNF-a, IL-1
Which is worse, warm shock or cold shock?
Warm shock
What is the supportive,
antimicrobial
Anti-inflammatory
anticoagulant
therapies of sepsis?
1. Supportive care
--IV fluids
--supplemental O2/ventilation
2. Antimicrobial
--Broad spectrum initially
--Directed abx once organism ID'ed
3. Anti-inflammatory
--Low dose hydrocortisone in pts who don't respond to fluids+vasopressors
4. Anticoagulant
--Activated Protein C
Describe the structure of EBV
dsDNA herpesvirus
What does EBNA2 gene do?
Transforms B cells into immortal B cells
What is the current antiviral treatment of EBV?
None effective
What mediates GVHD?
Donor T cells
Which graft sources are the lowest risk for histoincompatibility, which are the highest?
(most risky) PBSC > BM > Cord Blood (least risky)
Rank leukemias in order of easiest to cure with HSCT to hardest to cure.
CML > AML > ALL
What is the post-transplant immunosuppression regimen?
Calcineurin inhibitor (cyclosporin or tacrolimus) + methotrexate or mycophenolate
1. What is the most common viral infection after HSCT?
2. Why?
1. CMV
2. CMV reactivates during immunosuppression
Which myeloproliferative disorder leads to a shortened lifespan?
Myelofibrosis
What is the cure for myelofibrosis?
Allogeneic stem cell transplant
CD13 and CD33 are associated with which forms of leukemia?
Myeloid
CD5, CD7, and CD19 are associated with which forms of leukemia?
Lymphoid
What stain is used to identify ALL
Periodic Acid Schiff
1. What chromosomal abnormality is the best in childhood ALL?
2. What is the gene product?
t(12;21) - TEL/AML-1
What special precaution must be taken for ALL chemotherapy?
CNS prophylaxis
What are TDT and CD7 markers of?
Immature T-lymphocytes
How does Small Cell B lymphoma/CLL prognosis differ with ZAP 70?
ZAP70+ is worse
ZAP70- is better
1. t(11;14) is associated with what lymphoma?
2. What is the gene product?
3. What is the overall effect?
1. Mantle Cell lymphoma
2. Bcl-1/Cyclin-D1
3. hyperproliferation
1. t(8;14) is associated with what lymphoma?
2. What is the gene product?
3. What is the overall effect?
1. Burkitt lymphoma
2. C-myc
3. Hyperproliferation
A low power "starry sky" appearance is associated with what lymphoma?
Burkitt lymphoma
1. Bcl-6 point mutations or, rarely, t(3;14) are associated with what lymphoma?
DLBCL
What two chromosomal or genetic abnormalities are associated with DLBCL?
Bcl-6
t(3;14)
What is the treatment of DLBCL?
CHOP+R
What is the effect of Rituximab?
MAB to CD20
1. What lymphoma is associated with t(14;18)?
2. What is the gene product?
3. What is the overall effect?
1. FCC
2. Bcl-2
3. antiapoptosis
What lymphoma are butt cells associated with?
FCC lymphoma
1. Which lymphoma are clover leaf cells associated with?
2. What is the associated pathogen?
1. Adult T Cell Lymphoma/Leukemia
2. HTLV-1
1. What lymphoma is t(11;18) associated with?
2. What is the gene product?
1. MALT lymphoma (advanced)
2. API2
Which form of Hodgkin's lymphoma is most related to EBV?
Mixed-cellularity HL
t(4:14) is associated with what lymphoma?
multiple myeloma
1. Which thalassemia is prevalent among descendants of mediterranean families?
2. Which hemoglobins are elevated?
1. beta-thalassemia
2. Hb F (a2g2), Hb A2 (a2d2)
Which antigens are present on platelets?
Human Platelet Antigen (HPA)
HLA Class I
Blood Group A and B antigens
1. What is the function of gpVI?
2. What is the function of a2b1?
1. Binds collagen
2. Is expressed by platelet and stabilizes binding
What happens to platelets during cardiopulmonary bypass?
The platelets are activated in the mechanical pump and are thereafter dysfunctional
What is the effect of APC on PAI-1 and tPA/uPA?
APC inhibits PAI-1, thus de-inhibiting tPA and uPA
1. What does antithrombin+HMW heparin inhibit?
2. What does antithrombin+LMW heparin hinhibit?
1. Thrombin
2. IXa, Xa
What does HMW heparin+antithrombin inhibit?
Thrombin
What does LMW heparin+antithrombin inhibit?
IX
Xa
How is plasmin activity limited to the fibrin plug?
Plasmin binds lysines on fibrin
1. How is TAFI activated?
2. How does TAFI function?
1. TAFI is activated by thrombin-thrombomodulin
2. TAFI removes terminal arginine and lysine residues from fibrin, preventing plasmin from binding
What component must be present in an allergen to elicit an immediate hypersensitivity reaction (Type 1)?
Protein
Which interleukin mediates allergic asthma independently of IgE and eosinophils?
IL-13
What is the cause of Grave's disease?
Antibody activates TSH receptor
What is the cause of Myasthenia Gravis?
Antibody inactivates AChR
1. Which form of hypersensitivity is the tetanus toxoid booster reaction?
2. What is it called?
1. Type III
2. Arthus reaction
1. What are the symptoms of DiGeorge?
2. Which chromosome is fucked up?
C - cardiac dysfunction
A - abnormal facies
T - thymic aplasia
C - cleft palate
H - hypocalcemia
del22q11.2
Mutations in C5-C9 lead to a increased risk of infection with what organism?
Neisseria spp.
Mutations in the IFN-g receptor lead to an incresed risk of infection with what organism?
Atypical mycobacteria
Mutations in TLR3 receptor lead to an increased risk of infection which what organism?
Herpes simplex encephalitis
What 4 components mediate type I hypersensitivity reactions?
Th2, IgE, Mast Cells, Eosinophils
Which organisms are patients with CGD likely to be infected with?
S. aureus
E. coli
Apsergillus
Candida
What is the genetic defect of X-linked agammaglobulinemia?
Defective Bruton's tyrosine kinase
Defective Bruton's tyrosine kinase leads to which syndrome?
X-linked agammaglobulinemia
What is the genetic defect of X-linked HyperIgM?
Defective CD40L on T cells leads to no class switching
Defective CD40L on T cells leads to what syndrome? How?
X-linked HyperIgM since an absence of CD40L on T cells means there can be no class switching
What are the four types of rejection?
Hyperacute
Acute vascular
Acute cellular
Chronic
1. Describe hyperacute rejection
2. What is a cause of hyperacute rejection?
Occurs within hours due to preformed antibodies, such as ABO mismatch
1. Describe acute vascular rejection
2. What is a cause of acute vascular rejection?
1. Occurs in days to weeks, presents with vasculitis
2. Recognition of endothelial antigens
1. Describe acute cellular rejection
2. What is a cause of acute cellular rejection?
1. Occurs in days to weeks, presents with parenchymal necrosis and lymphoid infiltration
2. Cytotoxic and delayed hypersensitivity response to alloantigens
1. Describe chronic rejection
2. What is a cause of chronic rejection?
1. Fibrosis of the organ
2. Cytokines released in response to alloantigens
Myeloperoxidase deficiencies lead to impaired host defense to what organism?
Candida
What is Wiskott-Aldrich syndrome caused by?
What is Wiskott-Aldrich syndrome characterized by?
1. Mutations in WAS protein, involved in cytoskeletal function, leads to decreased antibody production
2. Eczema, thrombocytopenia, immunodeficiency, bloody diarrhea
What are the two effects of APC?
1. Deactivate FV and FVIII
2. Deactivate PAI-1