Block 1 Exam 4 Part 3 Flashcards
What does TGF-β regulate?
Inhibits proliferation by turning off pro-growth genes and promoting cell cycle arrest
Which cancers frequently have mutations in TGF-_ receptors or SMAD4?
Colon, stomach, endometrium, and pancreatic cancers
What is the role of PTEN?
A tumor suppressor that brakes the PI3K/AKT signaling pathway
What syndrome is associated with germline PTEN mutations?
Cowden syndrome
What is the function of the VHL gene?
Regulates cellular responses to oxygen levels by degrading HIF1_
Which syndrome is associated with VHL mutations?
Hereditary renal cell carcinoma
What is the role of the STK11 gene?
Regulates cellular metabolism
What syndrome is associated with STK11 mutations?
Peutz-Jeghers syndrome
What does APC regulate in colonic epithelium?
Negatively regulates the WNT pathway by promoting _-catenin degradation
What disorder is caused by germline APC mutations?
Familial adenomatous polyposis
What percentage of sporadic colon carcinomas have APC mutations?
About 70%
What role does E-cadherin play in epithelial cells?
Maintains cell adhesion and regulates contact-mediated growth inhibition
What cancer is associated with germline E-cadherin (CDH1) mutations?
Familial gastric carcinoma
What effect does loss of CDH1 expression have?
Increased invasiveness and WNT signaling
What proteins are encoded by CDKN2A?
p16/INK4a and ARF
What cancer is caused by germline CDKN2A mutations?
Familial melanoma
Which pathway does TGF-_ inhibit in normal tissues?
Cellular proliferation
What role can TGF-_ play in cancer?
Enhances immune evasiveness of tumors
What does PTEN regulate?
PI3K/AKT signaling
What disorder is associated with germline PTEN mutations?
Cowden syndrome
What does VHL encode?
A component of a ubiquitin ligase complex that degrades hypoxia-induced factors (HIFs)
What syndrome is caused by germline VHL mutations?
Von Hippel-Lindau syndrome
What is the Warburg effect?
Cancer cells prefer aerobic glycolysis over oxidative phosphorylation even with ample oxygen
How is the Warburg effect used clinically?
PET scans detect tumors by their high glucose uptake
Why do cancer cells rely on aerobic glycolysis despite it being less efficient for ATP production?
It provides intermediates for biosynthesis needed for cell growth
What is the main metabolic function of mitochondria in growing cells?
To generate intermediates for biosynthesis, not primarily ATP
What signaling pathway promotes glucose uptake and glycolysis in cancer cells?
Receptor tyrosine kinase/PI3K/AKT signaling
How does MYC support cell growth?
It upregulates glycolytic enzymes and glutaminase for biosynthesis
What is autophagy?
A state where cells cannibalize their own components for energy under nutrient deficiency
Why is autophagy often disabled in tumors?
It allows tumor cells to grow under poor conditions without triggering autophagy
How can autophagy benefit tumor cells?
It may allow tumor cells to become dormant and survive for long periods
What is oncometabolism?
A process where mutations in metabolic enzymes like IDH promote cancer
What happens when IDH is mutated?
It produces 2-HG, which inhibits enzymes that use _-ketoglutarate
What effect does 2-HG have on TET2?
It inhibits TET2, leading to abnormal DNA methylation
How does abnormal DNA methylation contribute to cancer?
It misregulates cancer genes, driving oncogenesis
What cancers are associated with mutated IDH?
Cholangiocarcinomas, gliomas, acute myeloid leukemias, and sarcomas
What is Warburg metabolism?
A pro-growth metabolism favoring glycolysis over oxidative phosphorylation
How is Warburg metabolism induced in normal cells?
By exposure to growth factors
Which proteins induce Warburg metabolism in cancer cells?
Oncoproteins like RAS, MYC, and mutated growth factor receptors
Which tumor suppressors oppose Warburg metabolism?
PTEN, NF1, and p53
What happens during autophagy?
Cells consume their components for energy under stress
How do cancer cells handle autophagy?
They may mutate to avoid it or corrupt it for nutrient supply
What is an oncometabolite?
A metabolite that alters the epigenome, leading to oncogenic gene expression
Which mutation leads to the formation of oncometabolites?
Mutated IDH
What are the two pathways leading to apoptosis?
Extrinsic (death receptor) and intrinsic (mitochondrial) pathways
What does the intrinsic pathway respond to in cancer cells?
Stresses like DNA damage, hypoxia, and metabolic disturbances
How do cancer cells evade apoptosis?
By mutations that disable key components of the intrinsic pathway
Which proteins promote apoptosis by permeabilizing the mitochondrial membrane?
Pro-apoptotic proteins BAX and BAK
What proteins inhibit apoptosis by preventing mitochondrial permeabilization?
Anti-apoptotic proteins like BCL2 and BCL-XL
What is the role of BH3-only proteins?
They neutralize anti-apoptotic proteins, promoting apoptosis
How does loss of TP53 function affect apoptosis?
It prevents upregulation of pro-apoptotic proteins like PUMA, allowing cells to survive
What is an example of cancer overexpressing BCL2 to evade apoptosis?
Follicular lymphoma with BCL2 translocation
What is the therapeutic approach for cancers with overexpressed BCL2?
Drugs targeting anti-apoptotic BCL2 family proteins
How is apoptosis initiated?
Through intrinsic or extrinsic pathways
What happens after the activation of apoptosis pathways?
Activation of caspases that destroy the cell
Which pathway is most commonly incapacitated in cancers?
The intrinsic (mitochondrial) pathway
What translocation causes BCL2 overexpression in follicular B-cell lymphomas?
(14;18) translocation
What effect does overexpression of BCL2 family members have in cancer?
Linked to cancer cell survival and drug resistance
What are the three critical factors contributing to the immortality of cancer cells?
Evasion of senescence,evasion of mitotic crisis,capacity for self-renewal
How many times can most normal human cells divide before entering senescence?
60 to 70 times
What happens to cells that bypass senescence but are not immortal?
They enter mitotic crisis and die
What is the role of telomeres in mitotic crisis?
Progressive shortening of telomeres leads to chromosome instability and cell death
What enzyme is responsible for maintaining telomeres?
Telomerase
How do cancer cells maintain telomere length?
By expressing telomerase or using alternative lengthening of telomeres (ALT)
What is self-renewal in the context of stem cells?
The ability of a stem cell to divide and produce at least one daughter cell that remains a stem cell
What are cancer stem cells?
Cells in a tumor that have the capacity for self-renewal and are responsible for tumor persistence
Why are cancer stem cells thought to be resistant to conventional therapies?
Because of their low rate of cell division and expression of drug resistance factors like MDR1
What are cancer stem cells?
Stem cell-like cells in cancers responsible for tumor persistence
How do cancer stem cells arise?
Through transformation of normal stem cells or genetic changes in mature cells
What two mechanisms allow cancer cells to have limitless replicative potential?
Inactivation of senescence signals and reactivation of telomerase
What is required for tumors to grow larger than 1-2 mm in diameter?
The capacity to induce angiogenesis
What is the function of angiogenesis in tumors?
It provides nutrients and oxygen and removes waste products
What molecular event activates the angiogenic switch in tumors?
Increased production of angiogenic factors or loss of angiogenic inhibitors
What transcription factor is stabilized by hypoxia to stimulate angiogenesis?
HIF1_
Which protein upregulates the production of VEGF in tumors?
Mutated RAS or MYC
What role does p53 play in angiogenesis?
It stimulates anti-angiogenic molecules and represses pro-angiogenic molecules
What is bevacizumab used for?
It neutralizes VEGF to block angiogenesis in cancer
What has been the limitation of angiogenesis inhibitors in cancer treatment?
They prolong life but usually only for a few months
What triggers angiogenesis in tumors?
Hypoxia through HIF1_ activation
Which factor does p53 stimulate to inhibit angiogenesis?
Thrombospondin-1
Which oncogenes upregulate VEGF expression to promote angiogenesis?
RAS, MYC, and MAPK
What is the role of VEGF inhibitors in cancer treatment?
They prolong the clinical course but are not curative
What is the major cause of cancer-related morbidity and mortality?
Invasion and metastasis
What must tumor cells overcome during the metastatic process?
Immune defenses and adaptation to a new microenvironment
What are the two phases of the metastatic cascade?
Invasion of ECM and vascular dissemination
What are the four steps of ECM invasion by tumor cells?
Loosening of cell-cell interactions, ECM degradation, attachment to remodeled ECM, and tumor cell migration
What is the first step in the invasion process?
Dissociation of cancer cells from one another due to alterations in intercellular adhesion molecules
Which adhesion molecule is often altered in epithelial tumors?
E-cadherin
What is EMT (epithelial-mesenchymal transition) and which transcription factors control it?
A process integral to metastasis controlled by SNAIL and TWIST
What is the role of matrix metalloproteinases (MMPs) in invasion?
MMPs degrade ECM, promote tumor invasion, and release factors like VEGF
What happens when tumor cells change how they attach to ECM proteins?
They demonstrate complex changes in integrins, promoting survival and migration
What is anoikis?
A form of apoptosis triggered by loss of adhesion to ECM
How do tumor cells propel themselves through degraded basement membranes?
Via locomotion stimulated by autocrine motility factors, ECM cleavage products, and paracrine factors
What protects circulating tumor cells from destruction?
Clumping with other tumor cells or blood elements like platelets
What are the three factors influencing where metastases appear?
Location and vascular drainage, tumor cell tropism, and escape from dormancy
What is the “seed-soil” hypothesis in metastasis?
Tumor cells adapt to favorable environments in certain tissues that promote growth
What are the four steps involved in tissue invasion by malignant cells?
Loosening of cell-cell contacts,degradation of ECM,attachment to novel ECM components,and migration of tumor cells
How are cell-cell contacts lost during tumor invasion?
By the inactivation of E-cadherin through various pathways
What enzymes mediate basement membrane and matrix degradation?
Proteolytic enzymes such as MMPs and cathepsins
What additional effect do proteolytic enzymes have on the ECM?
They release growth factors and generate chemotactic and angiogenic fragments from ECM glycoproteins
How can the metastatic site of a tumor often be predicted?
By the location of the primary tumor; tumors usually arrest in the first capillary bed they encounter
What is organ tropism in metastasis?
A tumor’s preference for specific organs due to expression of adhesion or chemokine receptors whose ligands are found at the metastatic site
Which genes are associated with epithelial-mesenchymal transition (EMT) in metastasis?
TWIST and SNAIL
What is immune surveillance?
It is the process by which the immune system scans the body for emerging malignant cells and destroys them
What role do CD8+ cytotoxic T cells play in tumor immunity?
They recognize tumor antigens and kill tumor cells
How do tumors evade immune detection by antigen-negative variants?
By selecting for subclones that lose expression of immunogenic antigens
What happens when tumor cells lose or reduce MHC expression?
They avoid detection by cytotoxic T cells but may still trigger NK cells
What are PD-L1 and PD-L2
and how do they help tumors evade the immune system?,They activate PD-1 on T cells, inhibiting T-cell activation and helping tumors evade the immune system
What role does TGF-_ play in tumor immune evasion?
It is an immunosuppressive factor secreted by tumors to inhibit the immune response
What is checkpoint blockade therapy?
A treatment that removes inhibitory signals (such as CTLA-4 or PD-1) on T cells, enhancing the immune response against tumors
What is the role of CD8+ CTLs in tumor immunity?
They recognize tumor antigens on MHC class I molecules and kill tumor cells
What types of tumor antigens exist?
They include mutated proto-oncogenes, tumor suppressor genes, overexpressed proteins, and viral antigens
How do tumors avoid immune detection?
They can lose antigen expression, reduce MHC molecules, or secrete immunosuppressive factors like TGF-_ and PD-L1
What is the link between immunosuppression and cancer risk?
Immunosuppressed patients have a higher risk of cancers caused by oncogenic DNA viruses
How do immune checkpoint inhibitors work?
They block tumor immune evasion mechanisms, making the immune system more effective against cancer
What are the main contributors to genomic instability in cancer cells?
Loss of p53 function, DNA repair factor mutations, and DNA polymerase mutations
What is the role of DNA mismatch repair proteins?
They correct replication errors, and defects in these proteins can lead to microsatellite instability
What syndrome is associated with defective mismatch repair genes?
Hereditary nonpolyposis colon cancer (HNPCC), also known as Lynch syndrome
How does UV radiation affect DNA
and how is it repaired?,It causes pyrimidine cross-linking, which is repaired by the nucleotide excision repair system
What is the significance of homologous recombination repair?
It fixes double-stranded DNA breaks and cross-links; defects increase cancer risk
What is the function of the BRCA genes?
They are involved in homologous recombination repair, and mutations lead to increased risk of breast, ovarian, and other cancers
How do DNA polymerase mutations contribute to cancer?
They cause loss of proofreading function, leading to an accumulation of mutations
What role does regulated genomic instability play in lymphoid cells?
It allows antigen receptor diversification but can cause lymphoid neoplasms through errors in recombination
What increases the risk of cancer development in persons with inherited mutations in DNA repair systems?
Increased genomic instability
What syndrome is caused by defects in mismatch repair
leading to colon carcinomas?,Hereditary nonpolyposis colon cancer (HNPCC)
What condition is associated with defective nucleotide excision repair and high skin cancer risk?
Xeroderma pigmentosum
What disorders are characterized by defects in homologous recombination DNA repair?
Bloom syndrome, ataxia-telangiectasia, and Fanconi anemia
Which genes are involved in familial breast cancer due to DNA repair defects?
BRCA1 and BRCA2
What happens in cancers with defective DNA polymerase proofreading?
Genomic instability leads to increased mutation rates
How do T and B cells undergo regulated genomic instability?
Through somatic gene rearrangements, which can cause lymphoid neoplasms
What type of reaction do cancers provoke?
Chronic inflammatory reaction
How can inflammatory cells promote tumor proliferation?
By secreting growth factors like EGF and releasing proteases that liberate growth factors from the ECM
How do inflammatory cells contribute to removing growth suppressors?
Proteases degrade adhesion molecules, removing barriers to growth
What protects tumor cells from anoikis?
Tumor-associated macrophages express adhesion molecules like integrins
How do inflammatory cells induce angiogenesis?
By releasing factors such as VEGF
How do inflammatory cells activate invasion and metastasis?
By releasing proteases that remodel the ECM and stimulating tumor cell motility
What factors help tumor cells evade immune destruction?
Soluble factors like TGF-_ and PD-L1 produced by macrophages
What are the most common chromosomal abnormalities in cancer?
Chromosomal translocations
How can translocations activate proto-oncogenes?
By promoter/enhancer substitution or by forming fusion genes
What is an example of a fusion gene in cancer?
BCR-ABL in CML
How can chromosomal deletions contribute to cancer?
By leading to the loss of tumor suppressor genes
What are two patterns of gene amplification seen in cancer?
Double minutes and homogeneous staining regions
What is chromothrypsis?
A chromosomal shattering event that creates multiple rearrangements
What are the types of oncogenic mutations found in cancer?
Point mutations, translocations, deletions, and gene amplifications
How do translocations contribute to cancer?
By overexpressing oncogenes or creating fusion proteins with altered signaling
What effect do deletions have in cancer?
They cause loss of tumor suppressor genes and can activate proto-oncogenes
How does gene amplification contribute to cancer?
It increases the expression and function of oncogenes
What is chromothrypsis?
A process where a chromosome is shattered and reassembled haphazardly
What is epigenetics?
Regulation of gene expression by mechanisms other than changes in DNA sequence
How can tumor suppressor genes be silenced in cancer?
By local hypermethylation of DNA
What effect do global changes in DNA methylation have in cancer?
They alter the expression of multiple genes
How do changes in histones affect cancer cells?
They modify gene expression that influences cellular behavior
What role does epigenetic therapy play in cancer treatment?
Drugs target enzymes responsible for reversible epigenetic modifications, like DNA methylation inhibitors and histone deacetylase inhibitors
What is initiation in chemical carcinogenesis?
It is the process by which exposure to a carcinogenic agent causes permanent DNA damage
What are promoters in chemical carcinogenesis?
They induce tumor formation in initiated cells but are not tumorigenic by themselves
How do promoters contribute to cancer?
They stimulate proliferation and clonal expansion of mutated cells
What is the role of unopposed estrogenic stimulation in cancer?
It acts as a promoter by stimulating proliferation of cells in the endometrium and breast
What is the nature of chemical carcinogens?
They are highly reactive electrophiles that react with DNA, RNA, or proteins
What is the difference between direct-acting and indirect-acting chemical carcinogens?
Direct-acting carcinogens cause cancer without metabolic conversion, while indirect-acting carcinogens require metabolic activation to become carcinogenic
What are direct-acting carcinogens?
They do not require metabolic conversion to become carcinogenic
What is a key example of a direct-acting carcinogen?
Alkylating agents used in cancer chemotherapy
What are indirect-acting carcinogens?
They require metabolic conversion to become active carcinogens
What are ultimate carcinogens?
They are the active carcinogenic products formed after metabolic conversion of indirect-acting carcinogens
What is an example of an indirect-acting carcinogen found in tobacco?
Benzo[a]pyrene
How are most indirect carcinogens metabolized?
By cytochrome P-450-dependent monooxygenases
What is a factor that influences susceptibility to carcinogenesis from indirect-acting carcinogens?
Polymorphic variants of P-450 genes that affect enzyme activity
What do most chemical carcinogens target?
DNA
What type of mutations do chemical carcinogens typically cause?
Mutations in oncogenes and tumor suppressor genes like RAS and TP53
What is a mutational hotspot associated with aflatoxin B1 exposure?
p53 gene at codon 249
Where is aflatoxin B1 commonly found?
Improperly stored grains and nuts
What type of cancer is associated with aflatoxin B1?
Hepatocellular carcinoma
What is a common mutation pattern in lung cancers caused by smoking?
Skewed base substitutions caused by carcinogens in cigarette smoke
What are some examples of other potential environmental carcinogens?
Vinyl chloride, arsenic, nickel, chromium, insecticides, fungicides, and polychlorinated biphenyls
What do chemical carcinogens typically damage?
DNA
What is the difference between direct-acting and indirect-acting carcinogens?
Direct-acting agents do not require metabolic conversion, while indirect-acting agents must be metabolized into an ultimate carcinogen
What influences the activity of indirect-acting carcinogens?
Polymorphisms of endogenous enzymes like cytochrome P-450
What role do promoters play in tumorigenesis?
They stimulate proliferation of mutated cells after exposure to a mutagen or initiator
What are some examples of direct-acting human carcinogens?
Alkylating agents used in chemotherapy
What are examples of indirect-acting human carcinogens?
Benzo[a]pyrene, azo dyes, and aflatoxin
What are examples of promoters that enhance tumorigenesis?
Agents causing hyperplasias of the endometrium or regenerative activity in the liver