Week 4 Flashcards

1
Q

AML Prognosis

A

o Cytogenetics = the single most important prognostic factor

• Favorable risk: t(8;21), t(16;16), t(15;17)
• Intermediate risk: normal karyotype (use FISH)
• NPM1+/Flt3- genotype most favorable

Unfavorable risk: del 5, del 7, trisomy 8, ***11q23 [common on prior exposure to chemotherapy], other complex karyotypes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

AML-M4

A
  • Hyperleukocytosis

* Hyperviscosity – sludging in vasculature with ischemia and/or infarct → blurred vision, HA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

CLL diagnosis (on test)

A

Flow cytometry: CD5+CD19+CD20+CD23+

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

AML clinical features

A

o 10,600 new cases in US in 2002 →7,400 deaths in US in 2002
o Median age diagnosis: 63 (80% >15)
o Increased risk with: Down Syndrome, Ataxia telangiectasia, Fanconi anemia, Li Fraumeni syndrome, Wiskott-Aldrich, familial leukemia, myelodysplasia, PNH,
o Secondary AML described with prior chemotherapy, radiation exposure, benzene (spill in Duluth area)
o Pancytopenia: anemia (often asymptomatic in elderly) + neutropenia + thrombocytopenia
o B symptoms: fever, night sweats, chills, malaise, weight loss
o Extramedullary disease: skin, CNS, orbits, bone, lung, kidney, bone, spleen, liver, ovary
• Monocytic leukemias most common → GUMS common (Dilantin can also cause this)
o Hyperleukocytosis: >100,000 blast count/ml
• APML, monocytic AML, inv(16), 11q23
• Tx promptly with hydroxyurea, leukopheresis, chemotherapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Acute Promyelocytic Leukemia (M3)

A

DON’T LET SUN SET ON M3
• Cytogenetics: makes a difference how you treat them!
o ***Most with t(15;17) – creates fusion gene, PML/RAR-alpha
o Poor risk disease with t(11;17)
• Treatment: induction therapy with ATRA plus anthracycline-based chemotherapy
o Consolidation with 2 courses anthracycline-based chemotherapy
o 2 years maintenance chemotherapy with ATRA, 6-MP, and methotrexate
o Relapse: arsenic trioxide
• DIC a common presentation: coagulopathy, depressed fibrinogen, thrombocytopenia → fatal hemorrhage
o If M3 and DIC, then desire to start ATRA (all-trans retinoic acid) in

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Mantle cell lymphoma

A

is a nasty, aggressive NHL: CD5+CD23- cyclinD1+ (don’t want to miss mantle cell)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

CLL cytogenics

A

ALWAYS get FISH studies with CLL

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

o Anergy

A

self- tolerance = T cell binds to APC without CD28 binding CD80/86 (B7) → T cell may be inactivated or die
• Makes sure the T cells being simulated are activated in an appropriate setting
• One way street – even if T cell encounters antigen later on in life, it will not respond

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

o Activation

A

TCR binds MHC + CD4/8 co-receptor + CD28 binds CD80/86
• IL-2 + high affinity IL-2 R produced → initiates activation → division, differentiation, effector functions
• Also need another signal to polarize T cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

o Activation Inhibited

A

= T cell binds APC but CTLA-4 binds CD80/86 instead of CD28 → TURN OFF SIGNAL
• CTLA-4 outcompetes CD28 to binding with CD80/86→ blocks co-stimulatory effect
• CD28 will also be on surface but CTLA-4 binds better and leads to inactivation
• Peak [CTLA-4] occurs 2-3 days after initial activation → prevents overstimulation of T cells
• Constant IL-2 release → constant proliferation of T cells BUT CTLA-4 ensures that you get a quick burst of T cell replication instead of runaway replication

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

b cell antigen presentation

A
  • Must be activated by Ag binding to Ab before they express co-stimulatory molecules
  • Present soluble antigens, toxins, and viruses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

o TAP

A

= transporter associated with antigen processing
• TAP complex has an affinity for peptides of 8-16 AA → optimal binding size of the MHC I is 9 AA
• Final trimming done in the ER by ERAAP (ER-associated aminopeptidase)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

invariant chain

A

CD74)
• Invariant chain assists in folding of the Class II α & β chains, binds to the peptide-presenting site of Class II molecules, & assists in transport of the MHC II molecules from the Golgi to cytoplasmic vesicles
o Proteolytic cleavage gradually digests invariant chain leaving a short fragment (CLIP) bound to antigen presenting site on the MHC II molecule (CLIP = Class II-associated invariant chain peptide)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

• Assembly of Class II MHC Molecules

A

o A nonclassical (HLA-DM) MHC II molecule is required to catalyze the exchange of antigenic peptide for the CLIP
• HLA-DM can be regulated by HLA-DO (DM promotes exchange, DO blocks exchange)
o HLA-DO is only expressed in B cells and in thymus, it is not induced by IFN-gamma
• Blocks HLA-DM except in very acidic conditions (late endosome)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

• MHC II Deficiency

A

o MHC II deficiency = autosomal recessive trait, health problems show up early in infancy (earlier than MHC I def.)
• Affected babies present with a mild form of combined immunodeficiency – increased susceptibility to pyogenic & opportunistic infections
• Differs from SCID – they have T cells which can respond to nonspecific T-cell mitogens such as PHA
o Patients with MHC II deficiency are deficient in CD4+ T cells, also have moderate to severe hypogammaglobulinemia
o Tx: hematopoietic stem cell transplantation (HSCT)
o Genetic linkage analysis → condition is NOT linked to MHC locus on Chr 6
o IFN-γ induces expression of MHC II molecules on APCs from normal people but fails to induce their expression on the APCs of patients with MHC II deficiency
• Lack of MHC II due to defects in transcription factors required to regulate their coordinated expression
o Pneumocystis jirovecii = opportunistic fungal infection, almost always associated with immunocompromised individuals
o WBC elevated – lots of neutrophils, low lymphocytes (normal is 40-60% neut, 22-44% lymph)
• 27% B cells (anti-CD20, HIGH), 47% T cells (anti-CD3) – 34% CD8, 10% CD5 (1:3 CD4:CD8 ratio)
• Substantial number of T cells + normal response to PHA → not SCID (no T cells/functional T cells)
o Lack CD4+ T cells – without IFN-γ there isn’t proper MHC II expression to expand CD4+ cell lines
o Low Ig levels because no CD4+ action to simulate them
o Patients reject skin grafts because they have fully functional CD8+ T cells, but it takes longer because they lack the initial CD4+ burst

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

o BCR complex

A

membrane-bound immunoglobulin (mIgM) + signaling chains CD79a and CD79b

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

• Pro-B Cells

A

earliest stage of antigen-independent B cell development
o Pro-B cells can be divided into 3 groups based on the expression of TdT and CD45R
• Early pro-B cells = TdT alone
• Intermediate pro-B cells = both TdT and CD45R
• Late pro-B cells = CD45R and have downregulated TdT
o CD45R – (R for cell growth & differentiation) remains expressed on surface throughout remainder of B-cell ontogeny
o As cells progress through pro-B cell stage, they rearrange their Ig heavy chain genes and begin to express CD43 (leukosialin), CD19 (BCR co-R, works with CD21 & CD81), RAG (recombination-activating gene)-1 and RAG-2.
o As late pro-B cells pass into the pre-B-cell stage → downregulate TdT, RAG-1, RAG-2, and CD43
o Pro-B cells also express c-Kit which binds to stem-cell factor expressed on bone marrow stromal cells
• Induces pro-B cells to proliferate and differentiate into precursor B cells (pre-B cells)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

• Pre-B Cells

A

o Pre-B cells express IL-7R and are stimulated to divide and differentiate using IL-7

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

• Immature B Cells

A

final stage of B-cell development in the bone marrow
o Immature B cells have successfully rearranged their light chain genes and express IgM
o Once again, RAG-1 and RAG-2 expression has been downregulated
o As immature B cells develop further into mature B cells, they begin to express both IgM and IgD on their surface
o These mature B cells are then free to exit the bone marrow and migrate into the periphery → transition phase

20
Q

• Cytokines Required for B Cell Development

A
o	Common lymphoid progenitors (CLP) are responsive to IL-7 which promotes B-cell lineage development
•	Mice deficient in IL-7 or IL-7R exhibit an early arrest in B-cell development at the pro-B cell stage
o	Blys (B-lymphocyte stimulator) signaling through its receptor BR3 is important for the survival of pre-immune B-cell stages from transition stage onwards
o	IL-4, IL-3 & low-molecular-weight B cell growth factor are important in initiating process of B-cell differentiation
21
Q

• Abnormalities in B Cell Development

A

o Immunodeficiency XLA → leads to a block at pro-B cell to large pre-B cell transition in the bone marrow
• Mostly caused by a mutation in gene encoding the enzyme Bruton’s tyrosine kinase (Btk)
• Btk = key E involved in signal transduction downstream of the pre-BCR and BCR
• XLA patients have very few circulating B-cells and negligible serum immunoglobulin (Ig)
• Accounts for ~85% of the cases of agammaglobulinemia
o Other causes of agammaglobulinemia are characterized by mutations in μHC, λ5, Igα (CD79a), Igβ (CD79b), and BLNK (SLP-65), all affecting pre-BCR functions
o Common variable immunodeficiency (CVID) impacts later stages of B-cell development
• CVID manifests in reduced serum Ig, memory B-cells, class switch recombination and B-cell activation
• Mutations in CD40 ligand on T-cells, B-cell surface receptor CD19, activated T-cell costimulatory molecule ICOS, and TACI (another receptor for Blys) have all been identified in CVID patients

22
Q

• B Cell Activation (T-Independent)

A

o T-dependent (TD) antigen: immune response depends on both T & B cells recognizing antigen in a linked fashion
o T-independent (TI) antigen: small number of antigens, can activate B cells w/o MHC II-restricted T cell help
o TI antigens can be divided into 2 groups (TI-1 and TI-2) based on the manner in which they activate B cells:
• TI-1 antigens: predominantly bacterial cell wall components –ex. lipopolysaccharide (LPS) on G- bacteria
• TI-2 antigens: predominantly large polysaccharide molecules with repeating antigenic determinants (ex. Ficoll, dextran, polymeric bacterial flagellin, and poliomyelitis virus)
o Many TI antigens are PAMPs that can be recognized by TLRs → activate B1 B cells
o B Cell response to Type 1 TI Antigen
• B1 B cells bind LPS through TLR4 or BCR
• TLR4 = nonspecific (polyclonal activation), BCR = specific (clonal activation)
• Type 1 TI antigens can stimulate both immature and mature B cells through the use of TLR4
• Only IgM is produced in response to this stimulation
o B Cell Response to Type 2 TI Antigen
• B1 B cells bind to Type 2 TI antigens through cross-linking of BCR [BCR = specific (clonal activation)]
• Type 2 TI antigens can only stimulate mature B cells through the use of the BCR
• Mostly IgM is produced in response to this stimulation
• CD4+ Th2 T cells can be involved to produce cytokine for a full B cell response that can include class-switching
o T-Independent Antigens Induce Poor Memory
• TI antigens predominantly activate the B-1 subset of B cells found mainly in the peritoneum
• These B-1 cells can be identified by their expression of CD5, which is induced upon binding of TI antigens
• In contrast to conventional B cells, B-1 cells have the ability to replenish themselves

23
Q

• Cytokines in B Cell Development

A

o Th1 releases IL-2 to B cell as it is dividing and IFN-γ to help with differentiation
o Th2 releases IL-4 to B cells as it is dividing and IL-4/5/6/10/13 to help B cell differentiate into mature plasma cell & memory B cells

24
Q

• Positive and Negative Selection in the Thymus

A

o Positive selection = restriction. Must recognize self-MHC molecules (intermediate). “Self-restricted T cells”. Negative selection = self-tolerance. Cannot respond to self-antigens. 98% of thymocytes do not mature into mature T cells.

25
Q

• T Cell Activation and Differentiation

A

o After antigen binding many genes are activated
o Immediate early = within 30 minutes à transcription factors c-Fos, c-Myc, c-Jun, NFAT, and NF-kB
o Early genes = within 1 to 2 hours à encode IL-2, IL2-R, IL-3, IL-6, IFN-γ, others
o Late genes = more than 2 days later à adhesion molecules

26
Q

• Clonal Anergy

A

o If T cell comes into contact with fixed APC (no CD80/86) → anergic genes transcribed
o No matter what happens later on in T cells life (even if it encounters normal APC), there will be no response

27
Q

CTL activation

A

o Ag-specific signal (transmitted by TCR upon recognition of proper peptide): MHC I complex presented by licensed APC
o Co-stimulator signal is transmitted by CD28:CD80/86 interaction between the CTL-P cell and the licensed APC
o IL-2 secreted by a Th1 or Th17 CD4+ or the CTL itself → proliferation and differentiation of antigen-activated CTL-P to a fully active CTL

28
Q

• Generation of Effector CTLs with CD4+ Help

A

o For optimal proliferation and memory generation of CD8+ T cells, CD4+ help is required
• APC has found antigen in environment → taken it up & broken it down → displayed it via MHC II
• Naïve Th0 binds MHC II with TCR and CD4 coreceptor → interaction at interface with CD28/B7→ Th0 activated to become effector cell
• T cell polarized to Th1 or Th17 → activated effector starts to produce CD40L
• When Th0 becomes polarized to Th1 it will produce IFN-γ → positive feedback to APC to produce more IL-12 and also drives CD8+ into effector cytotoxic T cell
• CD40L interacts with CD40 on APC → reverses instructional program → T cell gives instruction back to APC
• Interaction between CD40L and CD40 is required to tell APC to cross-present on MHC I
• Also gives instruction to APC on what CKs to release to stimulate effector cell function of CD8+
o Sequential: Th1/17 is released to complete its effector function and then CD8+ binds
• APC has been licensed → cross presents to MHC I → naïve CD8+ can bind antigen-MHC I complex → CD8+ produces IL-2 and high affinity IL-2 R (CD25)
• Ramp up response to strong pro-proliferative signal → clonal expansion
o Simultaneous: both CD4+ and CD8+ bound to APC at the same time
• More robust response – lots of IL-2 = bigger/rapid clonal expansion of CD8+ T cell

29
Q

o Naïve CTL-P

A

NO cytotoxic activity
• Does not express IL-2 or high affinity IL-2R (CD25) until after activation (no proliferation)
• Express high levels of L-selectin (tethering) and CC47 (chemokine R) – homing and retention in lymph node
• Express low levels of CD44 and LFA-1

30
Q

o Effector CTL

A

exhibits cytotoxicity (starts to produce perforin and granzyme)
• Synthesize IL-2 and express high affinity IL-2R (CD25)
• Expresses low levels of L-selectin and CCR7
• Express high levels of CD44 and LFA-1 – homing and retention at sites of inflammation

31
Q

o Memory CTLs

A

CTLs may not require Th1 CD4+ help to reactivate

• Requires only low levels of IL-2 (can be produced by activated CTLs) for memory CTLs to become mature effector CTLs

32
Q

• CTL Killing

A

o Binding of the Target Cell
• TCR-CD3 complex on CTL recognizes peptide:MHC I complex on target cell
• Takes only 10s from start of interaction for transcriptional changes to occur → very quick!
• LFA-1 on CTL binds to ICAMs on target cell
• Antigen activation converts LFA-1 from low affinity to high affinity state for better binding
• After 5-10 min, the LFA-1 returns to low-affinity state → dissociation of CTL from target cell
• Transient binding allows CTL to wipe out multiple target cells (CTL dissociation & recycling)
o Mechanisms
• Perforin (pore formation) and granzyme (cleave caspases → apoptosis) secretions = enzymatic process
• Granzymes are serine proteases – cleave Bid and procaspse-3 → apoptosis
• Fas ligand protein on the cell membrane surface = signal transduction process
• Membrane-bound FasL binds to Fas on membrane of target cells and initiates killing
• Activates death domains (FADD) that cleaves caspase-8 → apoptosis
• CTLs can also kill by TNF production and secretion

33
Q

Natural Killer Cells (NK Cells)

A

• NK cells make up 5-10% of circulating lymphocytes – level of CD56 determines function
o 90% blood NK cells are CD56low = most effective killers of target cells
o 10% are CD56high = release cytokines (IFN-γ) – NO granules
• NK cells play major role in killing virus-infected cells, IC pathogen-infected cells, and tumor cells
• Some NK cells can produce IFN-γ (among other cytokines)
o IFN-γ tilts immune response toward Th1 by inhibiting Th2 and inducing IL-12 production by macrophages and DCs
o IFN-γ can activate macrophages (M1, angry) and NK cells
• NK cell activity is stimulated by IFN-α, IFN-β, IFN-γ, TNF-α, and IL-15
• NK Cells and Early Defense Against Viruses
o PAMPs bind TLRs → type I interferons released
o Recruit NK cells at site of infection – look for aberrant cell surface molecules on those that are asking for help
o Virus titer increases until you see appreciable amounts of NK cells
• NK cells buy us time until adaptive immune response can kick in and virus-specific CTLs can fight infection

34
Q

• NK Cells vs. CTLs

A

o NK cells:
• Express NKRs & CD16 (FCγRIIIA) – recognizes IgG → mediate antibody dependent cytotoxicity
• Don’t need to be educated in thymus, don’t undergo rearrangement of R genes (don’t make TCR or CD3)
o NK cell killing is not MHC restricted (no CD4 or CD8 coreceptor binding requirement)
• Two different receptors competing for inhibitor or pro-activity signals in NK cell
o NK cells may have memory? → may be able to select and clonally expand certain populations
o NK cell killing is similar to CTL killing: FasL expressed on surface, perforin and granzyme released from granules
• TNF expressed on surface and secreted

35
Q

• NK Cell Receptors

A

NK cells have both activation and inhibition receptors
o NK cell R fall into two categories, but members of each group can either activate or inhibit NK cells
• Lectin-like receptors (NKG2 family) – binds to HLA-E &MHC- I-like molecules (NKG2D)
• Most receptors are activating
• BUT CD94-NKG2A = high affinity (HLA-E), inhibitory signal → trumps any activation signal
• Immunoglobulin-like receptors (KIR = killer cell Ig-like receptors)
• Bind to most MHC I molecules (HLA-A, B, and C)
• Most receptors are inhibitory → NK cell ignores target cell

36
Q

• Lectin-Like Receptors of NK Cells

A

o Heterodimer of CD94 + NKG2 – except for NKDG2D which is a homodimer
• CD94 associates via a disulfide bond
o NKG2A Receptor= inhibitory receptor, contains intracellular ITIMs (immunoreceptor tyrosine inhibitory motifs)
o HLA-E presents Peptides of Other MHC I
• Leader peptides from MHC I on loaded onto HLA-E in ER – requires TAP and tapasin
• Presented at cell surface for review by CD94 R on NK cells
• Only way HLA-E can get out to membrane is with peptide loaded
• Tells environment that the cell is okay because it’s still making MHC I so don’t worry!
• MHC I molecules present antigenic peptides from cytoplasmic proteins that have been transported into the ER → presented to TCR on CD8+ CTLs

o NKG2D Receptor = homodimer, recognizes stress on surface of cells
• Stress ligands produced in face of oxidative stress, rapid proliferation, heat shock, viral infection, etc.
• If NKG2D binds stress ligand in absence of inhibitory signal → activation signal to kill cell

37
Q

• The Leukocyte Receptor Complex

A

o LRC = 1 MB region, including the KIR, LAIR, LILR and SIGLEC, and CD66 (CEACAM) families, as well as other genes relating to NK cell function, such as the DAP adaptor proteins
o KIR locus = 150 kB – a number of different haplotypes found here, divided into ‘A’ and ‘B’ haplotypes
• A haplotypes contain few KIR genes, only one of which (KIR2DS4) is likely to be activating
• B haplotypes have many more KIR genes, including several activating KIR
• Multiple copies all producing slightly different KIR genes – recent evolutionary adaptation
• Duplicating/changing rapidly because they’re the types of proteins that are recognizing HLA-A/B/C (which have tons of allotypes within human genome)

38
Q

• Killer Immunoglobulin-like Receptors

A

o KIRs consist of 2-3 EC Ig superfamily domains
o Inhibitory receptors: long cytoplasmic tails that contain ITIMs (immunoreceptor tyrosine inhibitory motifs)
o Activating receptors: short cytoplasmic tails and a charged lysine residue (K) in their transmembrane domains, which allows them to associate with an ITAM–containing adaptor molecule

39
Q

• Opposing Signals of NK Cell Killing

A

o No killing if: MHC I binds inhibitor receptor (even if ligand binds activating receptor) – MHC I levels high
o Killing if: ligand binds activating receptor and nothing binds inhibitor receptor – MHC I levels low
o In a normal cell, it is no uncommon to transiently express stress ligand on surface → binds activating receptor on NK cell BUT MHC I-inhibitor receptor signal wins out
o Expression of activating and inhibitor receptors are co-dominant – can express from mother & father on same cell

40
Q

NK Cells and CTLs complement each other

A

o NK Cells: kill cells that mask the presence of foreign antigen on MCH I
• Also have to be activated to function – have to interact with inhibitory signal at least one time
• Want to make sure it has a repertoire of inhibitory signals that work
o CTLs: kill cells that express foreign antigen on MHC I

41
Q

Antibody-Dependent Cell Mediated Cytotoxicity

A

• Effector cells associated with ADCC: NK cells macrophages, monocytes, neutrophils, eosinophils
• Effector cells bind to antigen via antibody (through FcR)
• Killing is mediated by cytolytic E release by macrophages, neutrophils, and eosinophils
o TNF release by NK cells, monocytes, and macrophages
o Perforin release by NK cells and eosinophils
o Granzyme release by NK Cells
• Four Categories of Ab Effector Function
o Virus and toxin neutralization → prevents pathogen-host binding
o Opsonization → macrophage phagocytosis
o Complement fixation and formation of MAC → phagocytosis or lysis
o Antibody-dependent cell mediated cytotoxicity (ADCC) → NK-induced apoptosis
• Functions of Fc Receptors
o Degranulation
o Opsonization of bacteria and phagocytosis → digestion in lysosome
o Maintaining serum levels of antibodies
o ADCC
• FcR Signaling vs. BCR
o Evolutionary correlation between signaling cascades, but context of cell responding leads to wildly different outcomes
o In BCR → signaling leads to proliferation, survival
o In cells (macrophage) undergoing ADC → activation (production of CKs, enhancement of phagocytic activity) → attack

42
Q

Chronic Myeloproliferative disorders

A

o Malignant proliferation of myeloid cells (not blasts, but maturing cells) in blood, bone marrow
o Four disorders: CML, PV, ET, MF (chronic myeloid leukemia–predominantly neutrophils, polycythemia Vera–predominantly RBCs, essential thrombocythemia–predominantly megakaryocytes/platelets, Myelofibrosis–all cell lines proliferating)
o Occur only in adults
o Long course
• Disorders of stem cells–no differentiation block happens so the cells can mature out into mature cells (not blocked at blast stage etc.)
• Features common to all 4 disorders
• Occur only in adults
• Long clinical course
• ↑ WBC with left shift (mild to moderate left shift–mostly mature neutrophils with some less mature forms)
• Hypercellular marrow
• Big spleen b/c as marrow filled with malignant precursor cells, there is no room for normal cells to grow in the marrow–person will make hematopoietic cells wherever he/she can!
• May evolve into acute leukemia–as an end stage event (CML especially) that all for diseases can do.
• Mutated tyrosine kinases–all of these disorders! There are several different tyrosine kinases. Tyrosine kinases in hematopoietic cells receive growth signals. The mutation is an activating mutation so the tyrosine kinase is always on (no longer needs growth signals) so it leads to growth at all times–nothing tells it to stop anywhere along differentiation→mature cell!

43
Q

Chronic Myeloid Leukemia

A

• Things you must know!
• Neutrophilic leukocytosis
• Basophilia
• Philadelphia chromosome
• Three clinical phases
• ——————————-
• Most abundant cell: mature segmented neutrophil
• Basophilia–elevated basophil count! There is no other cause than CML
• No fat in marrow–Hypercellular, predominantly myeloid cells
• Laboratory findings in CML
• ↑↑↑ WBC (usually really high)–all malignant
• Neutrophilia with left shift
• Basophilia
• ↓ Hemoglobin→anemia
• ↑ Platelet count (at first)–because increase in megakaryocytes as well! Malignant platelets. Normal platelets deceased.
• ↓ LAP–old test, Leukocyte alkaline phosphatase (enzyme present in neutrophils)–if benign, LAP should be increased, in CML–neutrophils don’t make LAP so it will be decreased.
• Chromosome 9(part breaks off) and moves onto 22
• All the action happens on chromosome 22-Philadelphia chromosome
• Clinical findings in CML
• Symptoms
o Slow onset
o Fever, fatigue, night sweats
o Abdominal fullness
• Signs
o Big spleen
o Big liver (b/c making hematopoietic tissue there)

44
Q

Polycythemia Vera

A
  • High RBC (makes blood sludgy)
  • Different from secondary polycythemia
  • Thrombosis and hemorrhage
  • Jak-2 mutation (tyrosine kinase mutation)
45
Q

• JAK-2

A

• JAK-2 in PV (used for diagnosis)
• Normal JAK-STAT pathway
o Cell signaling pathway
o Important in many different cell types
o Pulls signals to the inside of cell and down into nucleus
• JAK-STAT in PV
o Mutated JAK-2: activity increased in PV–pathway is always turned on!
o Cells grow on their own
• Important for diagnosis and drug therapy→95 % of cases have this mutation!
• JAK = Just another kinase. 2 domains: active part, inhibitory domain (keeps other domain under control).
• Normally: JAK phosphorylates receptor and STAT comes in, Signal transducer and activator of transcription…binds to phosphorylated receptor, JAK phosphorylates STAT…whole thing moves down into nucleus to activate transcription)
• In PV–inhibitory domain does not work!

46
Q

• Treatment & Prognosis of PV

A
•	Treatment
o	Phlebotomy
o	Maybe myelosuppressive drugs
•	Prognosis
o	Median survival: 9-14 years
o	Death from thrombosis or hemorrhage
o	Leukemic transformation in some patients