1 / 122

Tumor (really “GROWTH”) suppressor genes

Tumor (really “GROWTH”) suppressor genes. TGF- β  COLON E- cadherin  STOMACH NF-1,2  NEURAL TUMORS APC/ β - cadherin  GI, MELANOMA SMADs  GI RB  RETINOBLASTOMA P53  EVERYTHING!! WT-1  WILMS TUMOR p16 (INK4a)  GI, BREAST BRCA-1,2  BREAST KLF6  PROSTATE.

emmy
Download Presentation

Tumor (really “GROWTH”) suppressor genes

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Tumor (really “GROWTH”) suppressor genes • TGF-β COLON • E-cadherin STOMACH • NF-1,2 NEURAL TUMORS • APC/β-cadherin GI, MELANOMA • SMADs GI • RB RETINOBLASTOMA • P53 EVERYTHING!! • WT-1 WILMS TUMOR • p16 (INK4a)  GI, BREAST • BRCA-1,2  BREAST • KLF6  PROSTATE

  2. TRANSFORMATION & PROGRESSION • Self-sufficiency in growth signals • Insensitivity to growth-inhibiting signals • Evasion of apoptosis • Limitless replicative potential: Telomerase • Angiogenesis • Invasive ability • Metastatic ability • Defects in DNA repair: “Spell checker”

  3. Evasion of APOPTOSIS • BCL-2 Overexpression • Seen in B-cell lymphomas of follicular type t(14;18)(q32;q21),dominant with IgH genes and overexpression of BCL-2 protein • P53 (Pro-apoptic gene) - Directly activates Bax (Bcl-2 protein) • Reduced levels of CD95/Fas • FLIP protein – Inhibits Caspase 8 • Loss of APAF1(Apoptosis activating factor-1) • Increased IAP (inhibitors of apoptosis)

  4. LIMITLESS REPLICATIVE POTENTIAL • TELOMERES determine the limited number of duplications a cell will have! • TELOMERASE, present in >90% of human cancers, changes telomeres so they will have UNLIMITED replicative potential

  5. TUMOR ANGIOGENESIS • Q: How close to a blood vessel must a cell be? • A: 1-2 mm • Activation of VEGF and FGF-b • Tumor size is regulated (allowed) by angiogenesis/anti-angiogenesis balance

  6. Proteases of tumors can induce production of angiogenic factors and inhibitors • Proteases act on ECM components (E.g. Collagen) releasing bFGF and VEGF • VEGF triggers the NOTCH Pathway

  7. INVADE & METASTASIS • 1. Invasion of Extracellular Matrix • 2. Vascular Dissemination • Invading the Extracellular matrix requires breaching the basement membrane  Intersitial Connective Tissue  Vascular Basement Membrane 

  8. Invasion of ECM • 1. Detachment ("loosening up") of the tumor cells from each other • Down-regulation of E-cadherin and Beta-catenin reducing cell’s adherance to each other

  9. Invasion of ECM • 2. Degradation of ECM, e.g., collagenase, etc. • Break down the basement membrane • Proteases – MMP (matrix metalloproteinases) • Releasing growth factors (e.g. VEGF) and angiogenic, growth promoting effects with cleavage of Type IV collagen for example • 3. Attachment to matrix components • Breakdown by MMP allows tumor cell receptors to bind ECM proteins and stimulate migration • E.g. MMP2 cleaves laminin and MMP9 cleaves collagen IV  Change in tumor cell receptor attachment to ECM proteins  Stimulates Migration

  10. Invasion of ECM • Migration of tumor cells • Cytokines ( autocrine factors) • Cleavage of the ECM components (Laminin, collagen) and growth factors (IGF I, II) that have chemotactic activity • Stromal cells around tumor also producehepatocyte growth factor-scatter factor promoting motility

  11. TRANSFORMATIONGROWTHBM INVASIONANGIOGENESISINTRAVASATIONEMBOLIZATIONADHESIONEXTRAVASATIONMETASTATIC GROWTHetc.

  12. Vascular Dissemination • Following intravasation, clumps of platelet-tumor aggregates are formed • Extravastion of this tumor emboli at distant sites  Attachment to the endothelium  Exits through the basement membrane • Followed by migration to lymphoid tissue using the CD44 adhesion molecule (expressed on T cells) • CD44 binds to hyaluronate allowing migration and towards metastic spread • *Most metastases occur in the first capillary bed available to tumor

  13. Other possible mechanisms explaining metastasis: • 1. Tumor cell adhesion molecule ligandsare expressed on the endothelial cell of target tissue • Target organ variation in endothelial cell expression of adhesion molecule ligands • 2. Chemokinereceptors are expressed in cancer cells that allowing binding of chemokines expressed at specific tissues • E.g. CXCR4 and CCR7 receptors expressed in breast cancer cells • 3. Chemoattractants – E.g. IGF 1, II, can be expressed at target tissues

  14. Breast cancer cells, how do they adapt and make their environment habitable? • - Osteolytic behavior in bone by osteoclasts • Tumor cells  PTHRP (parathyroid hormone-related protein)  Osteoblasts  RANK ligand  Osteoclasts • Osteoclasts break down the matrix releasing growth factors

  15. Development of Metastasis • One hypothesis suggest why only some tumors metastasize? • “Metastasis Signature”  Multiple abnormalities taking place in the early stage of development • Intrinsic properties, microenvironment (stroma), angiogenesis, resistance to immune defenders, and local invasiveness • Other hypothesis  Rare clonal variation that develop metastasis

  16. DEFECTS IN DNA REPAIR • 1. Mismatch Repair • 2. Nucleotide Excision Repair • 3. Recombination Repair

  17. DNA REPAIR GENE DEFECTS • DNA repair is like a spell checker • HNPCC (Hereditary Non-Polyposis Colon Cancer [Lynch]): TGF-β, β-catenin, BAX • XerodermaPigmentosum: UV fixing gene • Ataxia Telangiectasia: ATM gene • Bloom Syndrome: defective helicase • Fanconi anemia

  18. Hereditary Non-PolyposisColon Cancer • Mismatch Repair • Proofreaders disabled • Microsatellite fragments  1 to 6 nucleotide repeats • Creates instability • Dna mismatch repair gene affect cell growth indirectly, through mutations in other genes  TGF-beta, beta-catenin, Bax

  19. XerodermaPigmentosum • UV radiation exposure • Pyrimidinedimers • DNA damage with defect in nucleotide excision repair

  20. Autosomal Recessive Disorders • Defect in DNA repair by Homologous Recombination • Ataxia – Telangiactasia– neurological degenerative disorder, exposure to Ionizing radiation • Defective ATM gene • Bloom Syndrome - Ionizing radiation resulting in developmental defects • Defect in gene of chromosme 15  encodes helicase  dna repair through homologus recombination • Fanconi anemia – Chemotherapeutic agents causing damage • Bone-marrow aplasia BRCA1 and BRCA 2  Proteins take part in homologous recombination repair pathway • Mutation of BRCA2 gene  chromosomal breaks & aneuploidy

  21. STROMAL MICROENVIRONMENT • “Cross-Talk” between the tumor cells and ECM • E.g. Laminin degradation by MMP2  Stimulates tumor cell motility • E.g. Type IV collagen degradation by MMP9  Release of VEGF • Storage of inactive growth factors in ECM, proteases release the active form (E.g. PDGF, TGF-beta, bFGF)

  22. STROMAL MICROENVIRONMENT • Inflammatory Cells and fibroblasts • Cytokines released during inflammation  Induce survival and progression of tumor cells • Chemokines Chemotactic properties • Fibroblasts  Important role in tumor cells • Desmoplastic response, highly fibrotic , promoting growth

  23. Metabolism of Tumors

  24. WARBURG EFFECT • Otto Warburg, Nobel Prize discovery on the metabolism of turmors • Cancer cells rely on Aerobic Glycolysis, Warburg Effect, instead of the mitochondrial oxidative phosphorylation • Several Hypothesis – • 1. During angiogenesis, vessels are poorly formed, activation of H1F1alpha due to hypoxia  increases expression of enzymes of glycolysis, and vasculature • 2. Exposure to state of continous hypoxia and normoxia • 3. Mutations in oncogene and tumor supressors (p53, RAS, PTEN) stimulate metabolic changes

  25. Metabolic Alterations • In addition, tumor cells need to build and maintain its membrane, proteins, and organelles --- Anabolic Pathway • Lipid, nucleotide, proteins (amino acids), glucose (carbon) • Glutamine  Both glycolytic and anabolic pathway

  26. PI3K/AKT/Mtor pathway • Important in regulating normal cell functions including amino acid, glucose uptake • In tumors, mutation of tumor supressors and oncogenes --- Abnormality in pathway can occur favoring survival and proliferation of tumor cells • E.g. PTEN has a negative regulation of this pathway,

  27. Other tumor supressors that supress this pathway:- • Peutz-Jegher syndrome mutated gene LKB1 ( Liver kinase B1) • – Limits or regulates cell growth by AMP-dependent protein kinasemechanism – Negative regulator of mTOR pathway • Supression of LKBI (also called STK11)  Increase in anabolic metabolism of cancer cells • TuberousSclerosis mutated genes (TSC1 and TSC2) ----- Negative regulation of mTOR

  28. CHROMOSOME CHANGES in CANCER • TRANSLOCATIONS and INVERSIONS • Occur in MOST Lymphomas/Leukemias • Occur in MANY (and growing numbers) of NON-hematologic malignancies also

  29. Translocations ------------- Activate Proto-Oncogenes • 2 common mechanism of translocation: • A) Lymphoid Tumors --- Swapping or movement of segment of chromosome to another chromosome • B) Hematopoietic tumors --- Two chromosomes recombine to form hybrid fusion genes for production of chimeric proteins

  30. Burkitt Lymphoma:- t(8:14) (q24;q32) • Translocation at chromosome 8 (c-MYC) and 14 ( IGH gene) • MYC containing segment of chromosome 8 movement to to chromosome 14q32 at IGH gene • Overexpression of MYC

  31. Follicular Lymphomas:- t(14:18) (q32:q21) • Translocation at chromosome 14 (IGH gene) and 18 (BCL2) • Activation of BCL2

  32. Chronic Myeloid Leukemia (CML):- t(9;22) (q34;q11) • Translocation at chromosome 9 (ABL gene) and 22 (BCR gene)

  33. What is the Philadelphia chromosome? • Chromosome 22 that exchanges portion (reciprocal translocation) of the chromosome with Chromosome 9 • Fusion of two genesBCR-ABL producing protein with tyrosine kinase activity • Found in both CML & ALL (Acute Lymphoblastic Leukemia)

  34. Ewing Sarcoma • Also known as PNET (Primitive neuroectodermal tumor) • T(11:22) (q24;q12) FLI 1 gene and EWSR1 gene • Chimeric protein EWS-FLI 1 • Fusion of most genes encode for transcription factors

  35. GENE AMPLIFICATION • Amplification of particular chromosomal regions that contain the oncogenic sequence • Hallmark Features:- • Double minutes ---- Small, centric structures or chromatin bodies of extrachromosomaldna • Homogenous staining regions • Once a gene is amplified, these amplified regions can be inserted into new choromsome • Lack a normal banding pattern and appear homogenous (G-banded karyotype)

  36. Gene Amplification • N-MYC ---- Neuroblastoma • ERBB2 --- Breast Cancers • C-myc, L-myc, N-myc --- Small cell lung cancer • **Greater the amplification, poorer the prognosis

  37. Epigenetic Changes • Heritable changes in gene expression without changing the DNA sequences • Post-translational modfications of histones+ DNA methylation ===== Gene Expression • Cancer cells --- Hypermethylation within promoter regions and global DNA Hypomethylation • Tumor supressor genes --- hypermethlation --- Silenced • E.g. BRCA1, VHL, MLH1

  38. Epigenetic Changes • Genomic Imprinting: • Maternal or paternal allele of a gene or chromosome is modified by methylation and inactivated

  39. miRNAs • Non-coding, single stranded RNA • RNA-induced silencing complex • Post-transcriptional gene silencing • Downregulate gene expression by translational repression, and deadnylation • In cancer cells, increase oncogene expression or reduce tumor supression gene expression

  40. miRNAs • 1. Reduced miRNA (Inhibitor of oncogene translation) --------------- Overproductin of oncogene • 2. miRNA reduces tumor supressor gene

  41. Molecular Basis of Carcinogenesis

More Related