• 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

1/148

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;

148 Cards in this Set

  • Front
  • Back
nuclear receptors
bind esm that are hydrophobic that readily cross PM and are lipid soluble:

testosterone, progesterone, estrogen, mineral corticoids, cortisol, thyroid hormone, retinoids, and vit d

all transcription factors --> regulates DNA transcription

have a conserved DNA-binding domain (DBD or C domain), A-F domains:

A/B is variable
AF1 and AF2 are activation function domains
ligand binding domain (LBD, E),

D and F are highly variable --> D contains nuclear localization signal, some receptors don't have F
orphan nuclear receptors
nuclear receptors proteins with unknown ligand that binds but have same molecular structure
Inactive nuclear receptor
binding of inhibitory protein, inactive conformation
Active nuclear receptor
binding of ligand causes release of inhibitory protein, movement to nucleus, bind DNA, and binding of co-activators --> Tc
Early primary response to steroid hormone
steroid hormone ligan binds to nuclear receptor --> complex activates primary-response genes --> synthesis of proteins in primary response
Delayed secondary response to steroid hormone
Effect of cycloheximide primary response protien --> shuts of primary response genes and turns on secondary response genes --> secondary response proteins
steroid hormones can alsobind to membraine-associated receptors to elicit biological responses
estrogen, progesterone, etc..

by activating MAP kinase, cAMP, PKC, Calcium channels etc
Cell surface receptors
Ligand-gated channels (Ion-chaneel coupled receptors), G-protein-coupled receptors, Enzyme-coupled receptors
Ligand gated channels
ESM (often NT) binds to ion channel protein and opens or closes channel,allowing entry or exit of ions
G-protein- coupled receptors
spans membrane
involves receptor, g protein, and enzyme
ESM (a first messenger) actvated receptor and g protein, then enzyme --> 2nd messenger
enzyme-coupled receptors
involves ESM in form of a dimer
first activates catalytic domain of enzyme

OR activates enzyme by association of receptor via ESM
scaffolds
bring signaling proteins together to increase efficiency, precision, and speed
signaling protiens
are molecular switches, turn activity on or off
intracellular signaling cascades
organization of some protein kinases
Effect of ESM binding PM receptor
ESM binds PM receptor, activations of receptor, activity of proteins on, relayed from receptro to proteins bound by scaffold, generatio of 2nd messenger, 2nd messenger amplifies signal, signals bind to effectors, regulates Tc
Assembly of signaling complexes (scaffolds)
depends on presence of specific binding domains on signaling proteins
4 common binding domains in intracellular signaling proteins
PH = pleckstrin homology domain
PTB = phosphotyrosine-binding
SH2 = Src homology 2 domain
SH3 = Src homology 3 domain
PH domain
recognizes phophorylated inositol lipid (PIPII or PIPIII)
PTB and SH2 domain
recognizes phopho-Tyr domain
SH3 domain
recognizes proline-rich motif
GTP-binding proteins (G-proteins)
Binding of GTP switches protein from inactive to active state. G proteins contain intrinsic GTPase activity, hydrolyzing GTP to GDP switches off activity
Protein kinases and phosphatases
binding of ATP either turns on or off protein by phosphorylation. often organized into phosphorylation cascades, where one protein kinase then phosphorylates the next protein kinase
Phosphoryl activation switches
sometimes the binding of a protein to a phosphorylated residue (usually Tyr) via a PTB or SH2 domain is enough to activate a protein
as number of cooperating molecules to activate a response increases, percentage of maximum activation of target protein..
increases
G-protein-coupled receptors activate ..
heterotrimeric g-proteins
GTPase activity of G-protein alpha subunit is regulated by
SGR (regulators of G-protein signaling)
cAMP regulates cAMP-dependent protein kinase (PKA)
takes 4 cAMP to activiate regulatory subunit... catalytic subunit then phosphorylates CREB in nucleus to regulate Tc
G(alpha)q
activates phospholipase C-Beta

hydrolyzes PIP2 via phospholipase C-Beta to generate diacylglycerol and IP3 second messengers.

Diacylglycerol activates protein kinase C

IP3 goes to ER to release Ca2+ via IP3-gated calcium channels

All regulate muscle activity
Protein Kinase C (PKC)
2nd messenger made from activated diacylglycerol

Ser/Thr protein kinase

activated by diacylglycerol, calcium and membrane phospholipid.

It phosphorylates Tc factors, metaboli enzymes, and other proteins
Calcium
is a ubiquitous second messenger

triggers muscle contraction and secretion and regulates intracellular signal pathways
Calcium concentrations
in cytosol is LOW (10-7), so can act as a 2nd messenger. its conc is increased 10-20 fold when ER lets Ca out via IP3 gated channels

its conc is 10-3 in ER and outside cell
calcium pumps
in mitochondria, er, and PM
3 types of calcium channels that mediate ESM-stiumulated elavation of intracellular Ca2+
IP3-gated channels in ER

Voltage-dep channel in PM: opens in response to membrane depolarization

Ryanodine receptors in ER: activated with a rise in intracellular Ca2+ (ca-induced ca release)
with increasing amounts of vasopressin, calcium output....
increases concentration and frequency, not amplitude
Calmodulin
Binds Ca2+ allosterically and cooperatively!! (NOT covalently)

Calmodulin targets: PM Ca2+ pumps, calmodulin-dep protein kinases, and others

NOT an enzyme: it binds other proteins and alters their activity.

so it binds calcium, binds Ca pump on PM, allows release of calcium into extracellular space
calmodulin-protein kinase II
calmodulin activates cAM protein kinase II --> it is important in memory
G(alpha) i
inhibits adenylyl cyclase, decreases cAMP
G(beta, gamma) i
are activated by the G-protien-coupled receptor: acetylcholine receptor!!

activated G(beta, gamma) i activates K+ channels

acetylcholine has inhibitory effect on heart
G(alpha) olf
activates adenylyl cyclase in olfactory neurons, increases cAMP which then binds to cyclic nucleotide-gated cation channels bringing Sodium and calcium in and Cl- out...depolarizes neuron --> nerve impulse. regulates smell and vision!!

why humans can distinguish 10,000 distinct smells via olfactory neurons

the receptor is on surface of cilia on olfactory neuron. the odorant molecule is the ESM
amplification
one ESM generates lots of 2nd messengers!
how to turn off signals mediated by G-receptors/proteins?
phosphodiesterases, phosphatases, Ca2+ pumps

WAYS:
1. ligand dissociates off receptor
2. receptor "uncouples" from G-protein --> desensitization 'adaptation' cuz ligand is still bound but receptor is unable to signal
3. receptor internalized!
Enzyme-linked cell-surface receptors
cytosolic domains of these either has intrinsic enzyme activity or associates directyl with an enzyme. span membrane ONCE
Receptor tyrosine kinases
all have tyrosine kinase activity

activation by ESM
1. dimerization of inactive recptor tyrosine kinases
2. autophosphorylation (phosphorylate each other)
3. this phosphorylation alters conformation and increases kinase activity
4. phospho-tyrosines create high-affinity docking sites for binding of signaling proteins
5. trasient association of signaling complex`
Insulin and IGF-1 receptors bind ..
intermediate adaptor protein (IRS1)--> the activated insulin or IGF-1 receptor phosphorylates IRS1 --> docking sites for other signaling molecules
Phosphorylated tyrosines serve as docking sites for
proteins with SH2 domains!!
ie
GAP, PI 3-kinase, phospholipase C (PLC)
How to activate Ras
activate a tyrosine kinase receptor
1. bind Grb-2 to phosphorylated tryrones via SH2 domains
2. Grb-2 binds to SOS via SH3 domain
3. SOS (GEF) is brought into vincinity of inactive Ras and releases GDP from Ras and binds GTP on Ras to activate
4. GAP hydrolyzes GTP to GDP to inactivate Ras.
Activated Ras causes..
downstream Ser/Thr phosphorylaition cascade

ie. mitogen-activated protein kinase (MAPK) pathway
Mitogen-activated protein kinase (MAPK) pathway
activated Ras activates Raf, MEC, and Erk to regulate protein activity and gene expression. Mutations in Ras cause cancer. Activation of Raf, MEC, and ERK are produced by P of Ser/Thr AAs
Deactivation of Ras
Dissociateion from Receptor
Phosphatase: dephosphorylation of receptor
Accelerating GTP --> GDP
Receptor Tyrosine Kinases activate PI 3-kinases by
Regulatory subunit of PI 3-kinase binds to phosphorylated receptor via its two SH2 domains binding to the phosphorylated Tyrosines on the receptor. Binding alters the conformation of PI 3-kinase which activates it
Activated PI 3-Kinase
promotes phosphorylation of PIP2 --> PIP3

PIP3 binds to and activates proteins via their PH domains
Tyrosine kinase-associated receptors
receptor is associated with Jak (Janus Kinase)

ESM include the cytokines -- interferons, interleukins, erythropoietin, and select hormones, growth hormone, prolactin

binding of cytokind causes receptors to dimerize, associated Jaks are brought together to cross-P each other on tyrosines. Active Jak P's receptor, creating docking sites for STATs and other proteins
STAT
signal transducer and activators of Tc; latent Tc factor.

STATs dock to P tyrosines on tyrosine kinase-associated receptor via SH2 domain

STATs are then P'ed by Jaks
STATs dissociate from receptor and dimerize
Migrate to nucleus and other gene regulatory proteins, target gene Tc
Cytokine effects via Jak-STAT pathway via tyrosine kinase-associated receptors
immune response to viral infection
RBC production
Stimulates growth and production of IGF-1
Stimulates milk production
Immune response to vacterial infection
Receptor-like tyrosine phosphatases
Unclear what ESM is!?

Some receptors have no extracellular domains and are only intracellularly regulated via SH2 domains

All have intracellular tyrosine phosphatase domains
Receptor Serine/threonine kinases: TGFBeta family
regulate pattern formation (development, morphogens), proliferation, cell death, extracellular matrix formation

1. ESM binds to and activates Type II receptor
2. Type II recruits and P's and activates kinase activity of Type I receptor.
3. Type I receptor P's Smad3 or 2 protein
4. P'd Smad binds co-Smad4, and translocates it to nucleus where it activates target genes
Receptor guanylyl cyclases
activated by natriuretic peptides that dilate blood vessels

suluble guanylyl cyclase activated by NO, also generated from nitroglycerine

GTP --> via NO --> cGMP increase in heart

Regulates
PDE, ion channels, PK
Pathways that regulate proteolysis
notch receptors
regulated proteolysis of B-catenin
NF-kB signaling pathway
Notch receptor
regulates proteolysis
play major role in development of most tissues

nerves cell are developed from epithelial cells by producing Delta signal that binds onto Notch receptor on epithelium cell and inhibits these cells
Nerve cell development from epithelial cells
Nerve cell express Delta signal that binds to Notch receptor on adjacent epithelial cell,
2. gamma-secretin cleaves the notch receptor
3. Notch receptor tail migrates to nucleus to CSL protein to activate gene Tc
Alzheimers
mutation of presenilin-1 required for the final cleavage to activate Notch causes cleavage of B-amyloid precursor protein (APP) a membrane protein expressed in neurons. Peptide fragment is released into Extracellular space in brain and they for amyloid plaques to cause nerve damage
regulated proteolysis of B-catenin
requires regulated proteolysis

In absence of extracellular signaling molecule Wnt, B-catenin is P'ed and targeted for proteosomal degradation. --> regulates Tc

In presence of Wnt, Wnt binds to Frizzled receptor, leading to inactivation of GSK3Beta, and accumulation of unP'ed B-catenin B-catenin is stable and nmoves into nucleus and activates target genes. it functions as a co-activator of other TFs.
NF-kB
latent transcription factor sequestered in cytoplasm by an inhibitor IkB
NF-kB Pathway
requires the ubiquitylation and degradation of P'ed IkB in proteasome. NF-kB is freed and is a transcription factor!
TNFalpha trimer
Tumor Necrosis Factor is produced in stressful situations. this is the ESM for NF-kB...when NF-kB is bound to DNA, Tc occurs and produces inflammatory responses
Cell Cycle
M - mitosis and cytokinesis
G1 - "Gap" phase
S - "DNA replication"
G2 - Duplication
Events of Euk Cell Cycle
M Phase: Prophase, prometaphase, metaphase, anaphase, telophase

Interphase: G1, S, G2
Ways to study cell cycle
Yeast:
b/c proliferate in haploid state (1N)
easy to study mutations
divide quick
Cell-division-cycle (cdc) genes discovered in yeast
divide at low temps not high
way to identify crucial genes for cell cycle
What type of cells used to study cell cycle
tumor cells
normal tissues
immortialized cell lines
S phase recognized by H-Thy incorporation or artificial Thy analog brdU into newly synthesized DNA

--> labeling index: how many cells in S phase!!
How to measure amount DNA in cell
Flow cytometry. Dye that binds DNA during S phase
cell cycle control systems
clock, initiate stages in correct order, each stage triggered once, stages triggered in complete, irreversible manner, back-up mechanisms, sensitive to environmental condictions,
esm signals
Checkpoints in cell cycle
arrest cell cycle if previous step is not complete!

G2/M CheckPoint: Is all DNA replicated? Is environment Favorable?

Metphase-toanaphase Checkpoint: Are all chromosomes attached to the spindle?

G1 Checkpoint: is environment favorable?
Cyclin-dependent Kinases (Cdks)
control cell-cycle
They are cyclically activated, S/T protein kinases

activated by cyclin
cell cycle progression is regulated by P'tion of specific proteins by Cyclin/cdk

Cdk activty terminated by cyclin degradation
Cyclin
activates Cdk and directs Cdk to specific targets.

concentrations oscillate during cell cycle

Hi during G1, S, Mitosis,
Low during metaphase-anaphase and start of G1

G1 cyclin, G1/S cyclin, S cyclin, A cyclin, M cyclin
Cdk activation
binding of cyclin and P'tion by Cdk-activating kinase (CAK), a S/T kinase

Wee 1 inhibits Cdk by phosphorylating Tyr on Cdk

Cdc25 promotes Cdk by dephosphorylating the site Wee 1 P'ed!!
Cdk inhibitor proteins
inhibit Cdk activity by p27, p21, etc it binds cyclin/cdk
Cdk/cyclin inactivated by proteolysis
p27 is ubiquitinated by SCF after it is phosphorylated

G1 cyclin is degraded by being ubiquitinated by SCF

SCF is continually active during cell cycle it targets phosphorylated things
Anaphase-promoting factor (APC)
regulates proteolysis of Cdk

APC catalyzes ubiquitylation of S and M cyclins and other regulatory proteins (securin). APC is active late in mitosis by Cdc20 and by phosphorylation
Cells held in G1 by....
Cdk inhibitor proteins such as p21 and p27

Absence of cyclins

other inhibitors
Neurons and muscle cells cell division
contstant G0 state in which Cdk and cyclin expression is terminally off
Entry into S phase by...
Mitogens (growth factors)

activation of MAPK pathway
1. PDGF binds to receptor tyrosine kinase
2. activates Ras
3. Ras activates MAP kinase kinase kinase, MAP kinase kinase, MAP kinase, activating expression of Myc protein
4. Myc promotes expression of G1 cyclin, SCF subunit to destry p27, and E2F gene, all entry into S phase!
Cell proliferation regulated by Pi-3 kinase pathway!
generated Akt regulates cell division
stimulated by receptor tyrosine kinase or SOMETIMES GPCRs
Insulin or IGF-1 binding to IRS1 regulations cell division too
by generating Akt
How activated Akt leads to cell proliferation
phosphorylates p27 --> degredation
phosphorylates FOXO1 --> degredation

Myc binds to G1 cyclin gene, increased G1 cyclin, activation of G1/Cdk
Retinoblastoma (Rb)
Blocks progression into S phase. G1-cyclin/Cdk P's active Rb which release E2F in an active form. E2F binds to promotors of many genes that encode proteins for S phase entry (G1/S and S-phase cyclins.)
Inhibition of E2F by Rb releived by...
ESMs
Positive Feedback!
Rb
First tumor surpressor identified
p27 degredation
by G1/S- and S-cyclin
G1 Checkpoint
Block activity of G1/S cyclin/Cdk and S-cyclin/Cdk via DNA damage

IF DNA is damaged, protein kinases are made that activate p53.
1. p53 arrests cell cycle by producing p21 gene
2. p21 inactivates G1/S-Cdk and S-Cdk
3. p53 also targets DNA repair genes

Mdm2 can degrade p53
Mutations can occur in p53 and result in cancer
Activators of p53
DNA damage, hypoxia, ribosome levels, NT triphosphate levels, inactivating mutation of Rb
S-cyclin P'tion of ORC prevents...
reformation of pre-RC
Mitosis activated by
M-phase Cyclin/Cdk
M-cyclin increases during G2, resulting in formation of M-cyclin/Cdk. M-cyclin/Cdk is maintained in an inactive state until entry into mitosis

1. M-cyclin allosterically binds to Cdk
2. Cdk-activating kinase (CAK) and Wee1 phosphorylate M-Cdk but its still inactive
3. Cdc25 dephosphorylates the phosphate put on by Wee1 to create active M-cyclin/Cdk
G2 Checkpoint
delays entry into mitosis in response to incomplete DNA replication. Damaged DNA sends signal that prevents activation of Cdc25.
1. p53 to repair damaged genes
M-cyclin/Cdk P's specific proteins to
induce assembly of mitotic spindles
Ensure that replicated chromosomes attach to spindle
trigger chromosome condensation
Trigger nuclear envelope breakdon
triggger actin skeleton reorganization
Trigger reorganization of Golgi and ER
Principle stages of M phase
separate and distribute chromosomes to each daughter cell receives an identical copy of genome
Prophase
chromosome condensation mediated by m-cyclin/Cdk P'tion of condensins
Prometaphase
breakdown of nuclear envelope beings with M-Cyclin/Cdk mediated P'tion of nuclear lamina,

1. breaking down of nuclear envelope, MTs can interact with chromosomes
Metaphase
sister chromatids held together by cohesin proteins, aligned at equator.

kinetichore detect if sister chromatids are attached to spindles
Kinetichore
large protein complex that assemgles at centromeres: binds MTs and monitor MT attachment to Sister chromatids. important in checkpoint between metaphase and anaphase
Anaphase
sister chromatids are separated by shortening kinetichore MTs.

separation triggered by anaphase promoting complex (APC) that inactivates cohesin that attaches sister chromatids
APC role in mitosis
separation of sister chromatids by inactivating cohesin, triggers anaphase

1. inactive APC is P'ed by cdc20 and helps to degrade securin and activate separase to cleave cohesins
metaphase-anaphase checkpoint
are all chromosomes attached to spindle? APC will not be activated because Mad1 and 2 are recruited...will not detach sis. chromatids if they arent
Telophase
Nuclear envelope reassembles as a result of INACTIVATION of m-cyclin/cdk and action of phosphatases
APC promotes
degredation of M-cyclin and other regulatory proteins , allowing for dephosphorylation of lamins and condensin and disassembly of spindle in cytokinesis
Cytokinesis
spindle is disassembled, chromosomes decondensed
What happens in G1?
cells grow. Cyclin/Cdk is surpressed by absence of cyclin and/or presence of CDIs
how is cell growth regulated ?
by PI-3 kinase pathway --> Akt for cell survival, growth , and division.
apoptosis
programmed cell death, bone marrow and intestinal cells die each year, necessary during embryonic development
Atresia
99% of oocytes die by apoptosis only 450 are ovulated! of the 800,000
significance of apoptosis
atresia, cancer, neurodegenerative disease, heart attack/stroke, development, tadpole, mouse foot, autoimmune disease, irreparable DNA damage
Necrosis vs apoptosis
apoptosis:
cell shrinkage
Membrane integrity maintained
role for mitochondria and cyt c
No leak of lysosomal enzymes
characteristic nuclear changes
apoptotic bodies form
DNA cleavage
Activation of specific proteases
regulatable process
evolutionarily conserved
dead cells ingested by neighboring cells

Necrosis:
cell swelling
membrane integrity lost
no role mitochondria
leak of lysosomal enzymes
nuclei lost
apoptotic bodies do not form
No DNA cleavage
no activation of proteases
not regulated
not conserved
dead cells ingested by neutrophils and macrophages
caspase
involved in apoptosis

cysteine at active site . aspartic in target protein

cleaves itself, target proteins

caspase cascade:
1.initiator caspase -->amplification tightly regulated
2. executioner caspase
3. cleraves cytosolic proteins and nuclear lamins
first initiator caspase activated?
initiator caspases oligermerize w/in adaptor, activated when in proximity to each other
Two methods of caspase activation
Extrinsic (receptor mediated)
Intrinsic (mitochondrial
Extrinsic caspase activation
activation of cell-surface "death receptors"
1. ESM activation of death receptor
2. binding of intracellular adaptor molecule to death domain of death receptor
3. adaptor recruits and binds multiple copies of procaspase-8 that is autoactivated and is "initiator caspase",
4. downstream effector caspases activated
5. cell death

General: procaspases aggregate triggered by adaptor and become active
Instrinsic caspase activation
activated by
DNA dmaage, treatment of cells with chemotherapeutic agents, groth factor withdrawal, reactive ocygen species

Release of cyt c from mitochondria and initiation of caspase cascade regulated by Bcl2 family of proteins
extrinsic receptors
Fas

TNF-alpha
can either induce apoptosis or cell survival (NFkB)
intrinsic apoptosis pathway
cyt c released from mytochondria by some sort of signal, apaf1 adaptor binds cyt c, procaspases9 binds adaptor and aggregates, activation of procaspase and its released to initiate caspase cascade.
Bcl-2 proteins regulate cyt c release
either inhibit apoptosis or stimulate

Inhibit
Bcl-2, and Bcl-XL (4 BH domains)

Stimulate
BH123 (3 BH domains)
BH3 only (1 BH domain): inhibit Bcl2, activate BH123
Inhibitors of Apoptosis (IAPs)
prevent accidental apoptosis
bind to and inhibit active caspases
IAP antagonists
inhibits IAPs by binding to them --> allows apoptosis to occur
How are BH3 and BH123 activated
DNA damage and oxidative stress
p53 role in apoptosis
DNA damage activates kinases that promote phosphorylation of p53 which stabilizes p53 and produces p21 protein which arrests cell cycle in G1. this results in apoptosis via BH3 only and Fas ligand production
FOXO1 role in apoptosis
in absence of ESM, expression of FOXO1 causes apoptosis
Reduced apoptosis because:
mutated/inactive p53
overexpression of anti-apoptotic proteins: IAPs, Mdm2
Constitutively active anti-apoptotic Akt: P's FOXO1 to red;uce Fas ligand expression
Excessive expression of Bcl2 from chromosomal translocation
Cancer cells
refelct their origin
derive from a single abnormal cell
Clonal evolution
evaluation of chromosomes in tumor cells

ie chronic myelogenous leukemia : have leukemic white blood cells with "Philadelphia" chromosome translocation between long arms of 9 and 22
Epigenetic change exs
imprinting and x-chromosome inactivation
Most cancers are initiated by genetic change
shared abnormality in DNA sequence that distinguishes from normal cells. Mutagens that cause cancer are carcinogens
Environmental factors
70% carcinogens

largely environmental not genetic
Cyt P450
activates some carcinogens
Ames test
Potential mutagen + activating liver extract + cyt P450

ability to create revertants --> mutagenesis
Tumor initiators
do not themselves give rise to a tumor, but cause latent genetic damage (are mutagenic)

ie tobacco smoke
Tumor promoters
can cause cancer if they are applied after treatment with a tumor initiator. They are not mutagenic (do not dmage dna)
Phorbol Esters (TPA)
tumor promoter creates PKC
Viruses and cancer
alter dna directly or act as tumor promoter
Bacteria
ulcrs and stomach cancer (helobacter pylori
Onset of cancer
Single mutation not enough to cause cancer
development of cancer requires mutations in many genes
Most cancers are sporadic and have no basis in heredity
Development is slow
Progression requires successive roudns of mutation and natural selection
Unstable
depend on defective control of cell death or cell differentiation
cancer cells maintain/acquire telomerase activity

Mutations in BRCA genes --> ovarian and breast cancer

Mutations in Rb --> Retinoblastoma in young kids
Stem cells and tumor onset
stem cells fail to produce more stem cells --> tumor of transit amplifying cells

or

stem cells fail to differentiate --> tumor

tumor is benign unless mutation
Proto-oncogenes
normal genes that promote cell growth and mitosis

mutations in proto-oncogene increases activity of its product protein (gain-of-function) and is called an oncogene.

ie

Ras, EGF receptor (Her2), Myc, Bcl2

Cancer: DNA mutations that positively effect expression of oncogenes (activating mutations of proto-oncogenes)
Tumor Suppressor genes
inhibit cell division. usually TFs or intermediates in signaling pathays. Mutated tumor suppressor genes (loss-of-function) they drive cell towards cancer. Effect is ressicive and requires both genes to be effective.

ie

Rb, BRCA, APC, p53

Cancer: DNA mutations that negatively effect expression of tumor suppressor genes (inactivating mutations)
Mutations in APC...
cause stabilization of B-catenin which results in increased Tc of its target genes including Myc and G-cyclin

APC is tumor suppressor gene
Her2
oncogene derived from EGF receptor in which ESM is missing....constituitively active...breast cancer
Ras
Proto-oncogene...mutated in 1/4 of cancers
p53
tumor supressor gene involved in cell-cycle control, apoptosis, and genetic stability

mutated in 50% all cancers
p53 is activated by DNA damage, mdm2 is degraded and p53 binds to p21 gene. p21 binds to G1/Cdk arresting cell cycle
P53 also codes for BH3-only and Fas ligand
E-cadherin
tumor suppressor gene: promotes cell-cell adhesion in healthy cells and mediates contact inhibition
Multidrug resistance
cancer cells become resistant to chemotherapy
Gleevac
Abl-selective tyrosine kinase inhibitor used to treat chronic myelogenous leukemia