Molecular and Cell Biology of Tumors Mgr. Lucia Knopfová, PhD. Prof. RNDr. Jana Šmardová, CSc. Institute of Experimental Biology Faculty of Science MU Brno Bi9910en 3. Mitogenic signaling I Proto-oncogenes, oncogenes, signaling pathways, regulation of cell cycle Molecular and Cell Biology of Tumors (1) Sustaining proliferative signaling (2)  Evading growth suppressors (3)  Resisting cell death (4) Enabling replicative immortality (5) Inducing angiogenesis (6) Activating invasion and metastasis (7) Genome instability and mutation (8) Tumor-promoting inflammation (9) Deregulating cellular energetics (10) Avoiding immune destruction Hallmarks of cancer Hanahan D. and Weinberg R.A., Cell 144 (2011) 646-674 Is cancer disease of the cell cycle? •  Deregulation of cell cycle is key factor in cell transformation •  Deregulation of cell cyle is not the only factor in cancerogesis, it is usually not fully transforming Cell cycle progression M - G1 - S - G2 M – DNA copies are separated; condensed chromosomes G1 – DNA content: 2N S – DNA replication G2 – DNA content: 4N G0 – senescent or non-dividing cells Postmitotic phase – without re-entering cell cycle, linked with terminal differentiation Cell cycle regulation Term „cell cycle clock“ indicates that in a cell there is a molecular „device“ that integrates all signals coming from inside and from extracellular space and determines whether cell enters active cell cyle or quiscent state. Weinberg RA. The Biology of Cancer. Garland Science 2007 Key molecules regulating cell cycle Ø  Cyclin-dependent kinases (Cdk, Cdc; human - 13) •  Have catalytic and regulatory subunits, kinase activity is dependent on interaction with respective binding partners – cyclins •  Cyclins (human - 29) •  Their expression levels fluctuate during cell cycle •  They affect the specific phase of the cell cycle by activating respective CDK kinase, afterwards are quickly degraded •  Some have other functions not cycle dependent expression Ø  Inhibitors (CKIs, cyclin-dependent kinase inhibitors) •  Ink4 family of CKIs (p16, p15, p18, p19) – target mainly Cdk4 and Cdk6 •  Cip/Kip family (p21, p27, p57) – more promiscuous, often interfere with binding of cyclin to CDK Key molecules regulating cell cycle Restriction checkpoint •  mitogenic stimulation induces cell cycle progression •  antimitogenic stimulation blocks cell cycle •  Cell cycle is dependent of exogenous signals only during the part of G1 phase – until restriction checkpoint (or G1 checkpoint or Major checkpoint) Restriction checkpoint: key regulatory point in cell cycle pRB – trigger of cell cycle •  pRB in hypophosphorylated state blocks the transition through the restriction checkpoint: it interacts with TFs of E2F family (8 genes) and thus prevent transactivation of their target genes (proteins encoded by these genes are required for S phase) •  When phosphorylated pRB losses the ability to bind with E2F and thereby the passage through the restriction checkpoint is possible ⇒ Regulation of the restriction checkpoint = regulation of pRB phosphorylation E2F family of transcription factors •  8 genes + alternatively spliced variants + multiple transcription start sites •  regulation: transcriptional, translational, posttranslational, protein degradation, biding of co-factors, cellular localization Kent LN a Leone G. Nat Rev Cancer 19: 326-338, 2019 Key molecules regulating cell cycle Ø  cyclin dependent kinases (Cdk, Cdc; human - 13) •  Have catalytic and regulatory subunits, kinase activity is dependent on interaction with respective binding partners – cyclins •  cyclins (human - 29) •  Their expression level fluctuates during cell cycle •  They affect the specific phase of the cell cycle by activating respective CDK kinase, afterwards are quickly degraded •  Some have other functions not cycle dependent expression Ø  inhibitors (CKIs) •  Ink4 family of CKIs (p16, p15, p18, p19) – target mainly Cdk4 and Cdk6 •  Cip/Kip family (p21, p27, p57) – more promiscuous, often interfere with binding of cyclin to CDK Ø  pRB Ø  E2Fs Phosphorylation of pRB during cell cycle Extent of pRB phosphorylation is tightly controlled during cell cycle: during M/G1 transition all phosphate residues are removed from pRB, after restriction checkpoint the extent of pRB phosphorylation gradually increases with peak at M phase (pRB is phosphorylated at serine and threonine amino acids) Weinberg RA. The Biology of Cancer. Garland Science 2007 Cell cycle regulation Lundberg AS F a Weinberg RA, Eur J Cancer 35: 1886-1894, 1999 Regulation of pRB phosphorylation Phosphorylation of pRB •  Positivelly regulated by: - complexes of cyclins D (D1, D2 and D3) and CDK4/CDK6 - complexes of cyclin E and CDK2 •  Negativelly regulated by: - inhibitors of cyclin:CDK complexes: p21WAF1 and p27KIP1 - inhibitors of CDKs: p15INK4B a p16INK4A Mitogenic signaling leads to increased expression of cyclins D and donwregulation of protein levels of inhibitors Antimitogenic signaling leads to overexpression of inhibitor(s) and decreased expression of cyclins D What is damaged in transformed cells? •  Tumor suppressor pRB (retinoblastoma, ..) •  Tumor suppressor p16INK4A (melanoma, ..) •  protooncogenes cyclins D –  amplification or overexpression of cyclin D1 in more than 50% of breast cancer, HNSCC –  Chromosomal translocations including cyclin D1 locus in mantle cell lymphomas: t(11;14) •  „periferal players“ – components of mitogenic and antimitogenic pathways Amplification of cyclin D1 locus in head and neck cancer •  Example of detection of CCND1 amplification (11q13) in head and neck squamous cell carcinoma (HNSCC) by FISH. There are 3 to 5 copies per cell. •  Approx 1/3 of HNSCC have amplification of CCND1 and thereby enhanced expression level of cyclin D1. As result regulation of pRB phosphorylation is disrupted Weinberg RA. The Biology of Cancer. Garland Science 2007 Cyclin D1 and mantle cell lymphoma (MCL) MCL is characterised by t(11;14)(q13;q32) translocation which juxtaposes Ig heavy chain gene (IGH) sequences (enhancer) with the CCND1 locus, leading to up-regulation of cyclin D1 expression. Note: CCND1=BCL1=PRAD1 What is damaged in transformed cells? •  Tumor suppressor pRB (retinoblastoma, ..) •  Tumor suppressor p16INK4A (melanoma, ..) •  protooncogenes cyclins D –  amplification or overexpression of cyclin D1 in more than 50% of breast cancer, HNSCC –  Chromosomal translocations including cyclin D1 locus in mantle cell lymphomas: t(11;14) •  „periferal players“ – components of mitogenic and antimitogenic pathways Mitogenic signaling Classification of protooncogenes I I. Growth factors II. Receptors for growth factors III. Proteins Ras IV. Non-receptor tyrosine kinases V. Transcription factors This classification reflects steps of signaling pathways: 1. Binding of ligand (growth factor) with receptor 2. Activation of receptor protein kinase 3. Signal transduction to nucleus via cascade of protein kinases 4. Activation of transcription factor Schematic outline of signaling pathway Schematic outline of signaling pathway Signaling pathways: examples Growth factors and receptor tyrosine kinases (RTK) Signaling via tyrosine phosphorylation is used mainly in mitogenic signaling. Other signaling pathways use serine and threonine residues. Growth factors and receptor tyrosine kinases (RTK) Growth factors: polypeptides that are produced by cells and triggers signaling for either start or stop of proliferation, differentiation and survival by binding to their receptors at the cell surface •  Evidence for involvement of extracellular signaling molecules in cell transformation: factors secreted by cancer cells are able to induce neoplastic transformation in vitro Stimulation: •  autocrine – growth factor stimulates producing cell •  paracrine – growth factor stimulates neighbourging cells •  endocrine – stimulation of distant cells Signal transduction by RTK (EGFR) After ligation with signal molecues receptors form dimers (all but IGF receptor that is convalently bound homodimer) and autophosphorylation is induced. Phosphorylation induces conformation change – thus catalytic domain is opened and substrates may bind to the binding domain. Binding of the substrates is mediated mostly by SH2 and SH3 (Src-homology) domains. Weinberg RA. The Biology of Cancer. Garland Science 2007 Type of mutations affecting growth factors and RTK •  Overproduction of growth factors (→ autocrine stimulation) •  Amplification of receptor: (a) concentration of receptors at the cell surface is so high that they cluster randomly and dimerize without interaction with ligand; (b) extremely efficient capturing of the extracellular signal: cells with high levels of receptor are stimulated for cell division even though the concentration of signal molecule is low and normally insufficient to activate mitogenic pathway •  Structural changes of receptor: activation is not dependent on binding of ligand (point mutations, short deletions, shortened forms as results of gene fusions, etc.) Alterations of RTK v-ErbB has unlike EGFR shortened extracellular domain ⇒ it transduces mitogenic signal constitutively (without the interaction with ligand) (1984 – discovery of the structural similarity of EGFR and v-ErbB) Weinberg RA. The Biology of Cancer. Garland Science 2007 Alterations of RTK v-ErbB has unlike EGFR shortened extracellular domain ⇒ it transduces mitogenic signal constitutively (without the interaction with ligand) (1984 – discovery of the structural similarity of EGFR and v-ErbB) Weinberg RA. The Biology of Cancer. Garland Science 2007 Alterations of RTK Fusion of genes for RTK with other genes Constitutive dimerization of receptor as a result of gene fusion with a gene that normally form dimers or oligomers. Weinberg RA. The Biology of Cancer. Garland Science 2007 Structure and main classes of RTK Weinberg RA. The Biology of Cancer. Garland Science 2007 EGF family Family of EGFR receptors: erbB-1 (EGFR); erbB-2 (HER-2/ neu); erbB-3 (HER-3); erbB-4 (HER-4) Growth factors, that binds to EGFR receptors: EGF, TGF-α, amphiregulin (AR), HB-EGF (heparin-binding EGF-like growth factor), cripto, betacellulin (BTC), epiregulin EGF family EGF (epidermal growth factor) - 53 amino acids - First identified and purified growth factor (Stanley Cohen – Nobel prize 1986) - Produced as a precursor (1217 AA) - Stimulates proliferation and differentiation of various cell types TGF-α (transforming growth factor α) - 50 AA, 40% homology with EGF, also mitogen - First identified autocrine growth factor - Produced as a membrane bound precursor, released by proteolytic cleavage in two steps - Expressed by many cancer cell lines and in many tumors that express EGFR EGFR family EGFR (EGF receptor) •  Is encoded by c-erbB (ERBB) gene – homologous with v-erbB (carried by retrovirus avian erythroblastosis virus: viral counterpart has deleted extracellur domain) •  Sometimes denoted as erb-B receptors or HER (human EGF receptor) •  Function as homodimers and heterodimers - Overexpression of EGFRs detected in breast, esophagus, stomach, ovarian, endometrial, cervical and other cancer; associated with poor prognosis in breast, esophagus and ovarian cancer -  Point mutations of EGFR1 (ERBB1) in lung cancer : EGFR - target of anti-cancer therapy by small inhibitors of kinase domain (TKI), point mutations predict good response to gefitinib EGFR family HER2/Neu encoded by gene ERBB2 Ø  amplification and/or overexpression of c-erbB2 (ERBB2) found in breast adenocarcinomas, stomach, salivary glands, ovarian and colorectal cancer Ø  In breast and ovarian cancer is amplification/overexpression of HER2/Neu associated with worse prognosis Ø  Amplification of HER2/Neu is prognostic and predictive marker in breast cancer: patients with HER2/Neu amplifications undergo more radical treatment and may receive Herceptin – monoclonal antibody specific for protein HER2/Neu Amplification of HER2/neu in breast cancer •  Amplification of HER2/neu first described in breast cancer in 1987 •  Amplification resulting in minumum 5 copies of HER2/Neu gene per cell is associated with poor prognosis Weinberg RA. The Biology of Cancer. Garland Science 2007 EGFR-targeted anti-cancer therapy A.  Monoclonal antibodies Herceptin (Trastuzumab) – anti ErbB2 (HER2): therapy of metastatic breast cancer with overexpression (amplification) of HER2/neu Cetuximab (Erbitux) – anti EGFR: therapy of advanced CRC positive for EGFR (Cetuximab binds EGFR with approx 50times higher affinity compared to EGF – mechanism of action is direct competition with ligand) K-ras status needs to be determined! (Why?) EGFR-targeted anti-cancer therapy B.  Small molecules – inhibitors of TK domain Gefitinib (Iressa), Erlotinib (Tarceva) – block ATP binding site of EGFR: therapy of advanced non-small lung carcinomas; gefitinib is efficient only in a subgroup of patients with specific mutations of TK domain of EGFR Lapatinib (Tyverb®) – 4-anulinochinazolin - dual inhibitor of HER1 and HER2 - inhibits intracellular tyrosinekinase domains of HER1 and HER2 - Used as a therapy of advanced metastatic breast cancer with overexpression of HER2 PDGF family PDGF (platelet-derived growth factor) – efficient mitogen – stimulates mesenchymal cells (fibroblasts, adipocytes, endothelial cells, smooth muscle cells) -  homodimer (AA, BB) or heterodimer (AB) of two related chains A and B: B is encoded by c-sis proto-oncogene -  In 1983 the homology between B chain and v-sis oncogene transduced by Simian sarcoma virus: altered B chain is constitutively expressed that leads to the sustained stimulation of cells with PDGF receptor -  Elevated expression of PDGF A or B or both frequently detected in sarcomas, gliomas, lung, breast, esophagus, stomach and colorectal carcinomas VEGF family VEGF (vascular endothelial growth factor) / VPG (vascular permeability factor) •  Mitogenic effect on endothelial cells, important for angiogenesis •  VEGF/VPG has 18% sequence homology with PDGF A and B FLT (FLT1 =Fms-Related Tyrosine Kinase 1, VEGFR1) encodes receptor for VEGF/VPG, binds VEGF with high affinity and mediates its biological function other receptors for VEGF described: KDR (VEGFR2) and FLT4 (VEGFR3, modulates KDR signaling by forming heterodimers) receptor encoded by FLT3 binds FLT3L not VEGF, expressed by haematopoetic stem cells, mutations in AML) FGF family •  Large family –many polypeptides: FGF1 to FGF18, rather original names are used (not all of them stimulate fibroblasts for proliferation!!) •  bFGF (FGF2) a aFGF (FGF1) (basic / acidic fibroblast growth factor) •  approx 17 kDa, they have 55% homology, similar function, name derived from different isoelectric points •  Mitogens of mesenchymal, neuroectodermal and epiderimal origin, involved in angiogenesis •  Autocrine growth factor in gliomas and meningiomas •  Overexpression found in advanced astrocytomas FGF family FGF receptors - 4 genes: FGFR1 (flg nebo fms-like gen), FGFR2, FGFR3 and FGFR4 – in addition genes can be alternativelly spliced, resulting in complex group of receptors (almost 100 combinations) with different affinity to FGF factors K-sam –FGFR2 -  Overexpression correlates with invasiveness and poor prognosis of stomach cancer, amplifications in breast and lung cancer: tumors with over-active FGFR signaling treated with targeted therapeutics: ATP competitive TKI inhibitors dovitinib, ponatinib (also inhibits mutant FGFR2) HGF family HGF (hepatocyte growth factor; hepatopoietin A) •  Stimulates proliferation of hepatocytes (not exclusively), involved in liver regeneration •  Is one of the scatter factors (SF): participates in loosening of the cell-cell interactions in epithelial tissues and blood vessels. c-met (MET) encodes receptor for HGF •  Amplification of c-met (stomach carcinomas, melanomas) •  mutant oncogenic form of c-met, constitutively active (without binding of HGF): point mutations, shortened extracellular domain; gene fusions •  HGF produced by stromal cells (overexpression) and c-met is expressed on stromal and cancer cells: cooperation in growth and morphogenesis of some tumors Oncogenic alteration of MET Comoglio et al. Nat Rev Cancer 18: 341-357, 2018 Hereditary papillary renal cancer (HPRC) •  autosomal dominant syndrome; highly penetrant •  Caused by inherited mutations of c-met; (locus 7q31) •  Papillary renal carcinoma represents cca 15 % of kidney cancer •  Predispose to multifocal, bilateral papillary renal tumors, not early metastatizing •  Not accompanied by congenital developmental defects •  Most germinal (as well as somatic) mutations are point missense mutations in TK domain of MET •  Met activation blocks terminal differentiation of renal tubular cells •  protooncogen c-met is mutated in more than 10 % of sporadic papillary renal cancers RET •  orphan receptor •  potential ligands: GDNF (glial cell line derived neutrotrophic factor), neurturin, persephin, artemin •  Highest similarity with FGFR •  Expressed mainly in neural, neuroendocrine and nephrogenic tissues Ø  2 types of mutations described: •  loss-of-function mutations (inactivation of RET receptor) – associated with congenital disorder Hirschprung‘s disease (absence of some or all ganglion cells in myenteric and submucosal plexus of colon, causing chronic constipation) •  gain-of-function = oncogenic mutation – activation of RET Hirschprung‘s disease •  1886 – Harald Hirschprung described the disorder causing death of two infants •  Absence of inervation leads to insufficient peristaltics and chronic constipation •  appears in 18.6 per 100,000 of live births •  Combination of mutations in two genes RET (CH 10) and EDNRB (CH 13) accounts for most cases •  RET receptor assists cells of the neural crest in their movement through the digestive tract during the development of the embryo •  EDNRB connects these nerve cells to the digestive tract Mutations in these two genes could directly lead to the absence of certain nerve fibers in the colon •  nearly 20% of affected children have associated developmental disorders and syndromes: often associated with Down syndrom (7%), deafness, medullary thyroid carcinoma, MEN2, neurofibromatosis, neuroblastoma •  Most cases are sporadic, familial burder increases risk 200x •  Multigenic disease with more than 10 involved genes, RET proto-oncogene is the most studied •  RET mutations: half of hereditary and 7-35% of sporadic cases Hirschprung‘s disease Ježová M et al. Cesk Patol 55 (1): 53-59, 2019 RET •  gain-of-function = oncogenic mutations gain-of-function is caused by several types of mutations: §  germline missense mutations •  Cause constitutive activation of RET •  Linked with hereditary cancer syndromes with increased malignancy risk (autosomal-dominant inheritance): MEN type 2A and 2B (multiple endocrine neoplasia) and FMTC (familial medullary thyroid carcinoma) Multiple endocrine neoplasia type 2 (MEN2) •  Autosomal dominant inheritance associated with mutations of RET •  gen locus: 10q11.2 •  type A: high risk of medullary thyroid carcinomas, often combined with feochromocytomas or benign adenomas of the parathyroid glands •  type B: Additional features include mucosal neuromas of the lips and tongue. •  type A is more frequent compared to type B •  Medullary thyroid carcinomas occur at different age - in 70 years there is a 70% penetrance Multiple endocrine neoplasia type 2 (MEN2) Pheochromocytomas •  tumor of the adrenal medulla (80%), approx 20% outside of adrenal gland (i.e. paraganglioma) •  90% benign, approx 10% malignant-metastatic •  These neuroendocrine tumors are capable of producing and releasing massive amounts of catecholamines, metanephrines, without treatment it is a life-threatening disease („malignant“?) •  Mostly sporadic •  around 20% hereditary, in this case multifocal; occur as a part of cancer syndroms: MEN II (A, B), FMTC, VHL, NF1 Familial medullary thyroid carcinoma FMTC •  Less frequent disease compared to MEN, manifestation later in life and less aggressive •  Associated with specific RET mutations RET mutations in MEN2 vs. FMTC 1.  Mutations occur mostly in extracellular part of receptor in cystein rich domain: C609, 611, 618, 620, 634 (in MEN 2A and FMTC) causing permanent dimerization (by disulfidic bonds between cystein residues) thus activation of receptor without ligand binding 2.  In some FMTC families mutations found in catalytic - kinase domain (codons 768, 804). 3.  MEN 2B syndrom is associated with mutations in second part of kinase domain (codons 883, 918) RET mutations in MEN2 vs. FMTC RET •  gain-of-function §  missense mutations §  Translocations resulting in deregulation of chimeric protein; RET discovered in 1985 as a proto-oncogene that underwent rearrangement during the transfection of DNA extracted from human T-cell lymphoma into NIH-3T3 cells (RET – REarranged during Transfection) Ø  Translocation of 3´- terminus of RET with 5´- part of other gene occurs in thyroid papillary carcinomas: chimeric protein contains tyrosine kinase domain of RET and polypeptide that allows permanent dimerization Ø  radiation-induced human papillary thyroid cancer (PTC) is associated with chromosomal inversions that involve the genetic loci H4 and RET on chromosome 10 RET RTK: Kit •  Originally identified as an oncoprotein endoced by feline sarcoma retrovirus •  Ligand for this receptor is SCF (stem cell factor); stimulates some blood cells but also nonhaematopoietic cells (e.g. melanocytes) •  Mutant forms of Kit transduce the signal constitutively, without ligation, typical mutations occur in juxtamembrane cytoplasmatic domain Weinberg RA. The Biology of Cancer. Garland Science 2007 Activation of Kit in cancer cells •  juxtamembrane (JM) domain negatively regulates Kit receptor •  Binding of ligand leads to transphosphorylation of tyrosine residues of JM domain. That opens JM domain to interact with Nterminal part of catalytical domain resulting in phosphorylation (activation) of kinase domain •  Mutations in JM domain result dimerization of Kit receptor, but receptor activity may be inhibited by TKI (unlike receptors with mutated catalytic domain) Sheikh E et al. Bosn J Basic Med Sci, 22(5):683-698, 2022 Mutation of c-kit •  GISTs (gastrointestinal stromal tumors) originate in mesenchymal cells of lower gastointestinal tract: normally function to ensure peristaltic contractions of smooth muscle cells (experimental mutations of Kit in these cells prevent this function in mice) •  activating mutation of c-kit found in almost all GISTs, frequently found in leukemias as well; often point mutations of JM domain, sometimes deletions of JM domain (worse prognosis) and mutations in catalytic domain. •  (hereditary inactivating Kit mutations – piebaldisms – absence of melanocytes in certain areas of skin and hair) Familial gastrointestinal stromal tumors (GISTs) •  hereditary GISTs associated with activating c-kit mutations in germinal lineage •  Set of germinal mutations is similar to somatic mutations occuring in sporadic GISTs •  Gen occupies locus 4q11-12 •  Example of a family with germinal Kit mutation: •  Deletion of 1 AA in JM domain •  Manifestation of the mutation late in life, thereby present in 4 generations Signaling pathways activated by RTK Signaling pathways activated by RTK Ras proteins ras/Ras – first identified oncogene Originally thought to function as mediator of Raf-1 binding to plasmatic membrane Current knowledge show that: •  Raf-1 activation is a complex multistep process. Direct interaction between Ras and Raf-1 is mediated by several binding sites in Ras and at least 2 binding sites of Raf-1 •  Raf-1 is not the only substrate (downstream effector) of Ras proteins. •  Ras superfamily consists of 35/65 different members (RAW/ 2000). All have some sequence homology, but differ in signaling capacity. Proteiny Ras Signal transmission from RTK to Ras •  Even transmission process is more complex than originally estimated •  activated RTKs recruit the adaptor protein GRB2 that binds phosphotyrosine consensus sequences via its SH2 domain. Via its SH3 domains, GRB2 then recruits the guanine nucleotide exchange factor SOS to the plasma membrane, where it activates the membrane-localized small G protein RAS by exchanging GTP for GDP Weinberg RA. The Biology of Cancer. Garland Science 2007 Ras proteins Ras superfamily has at least 65 members: Families: Ras, Rho, Rab, Ran, Rad, Arf Ras family has subfamilies: Rap, Ral, RRas, Rheb, H-Ras, Ras Ø Together called small G proteins Proteins H-Ras, N-Ras, K-Ras4A and K-Ras4B have 188/189 AA. They reside at the cytosolic side of plasma membrane and function there as molecular switches during signal transduction to cytoplasm. Ras proteins are GTPases: are inactive in GDP-bound form and active if GDP is exchanged for GTP. Capacity of Ras proteins to exchange of GTP/GDP and hydrolyse GTP is small, thereby other proteins participate: •  Guanine nucleotide exchange factors (GEFs), facilitate formation of active GTP-bound Ras protein, help to GDP to dissociate from small GTPase (e.g. SOS1/2, RasGRF/ mCDC25,...) •  GAPs: GTPase-activating proteins, act antagonistically to inactivate GTPases by increasing their intrinsic rate of GTP hydrolysis (p120 GAP, NF1/neurofibromin,...) Ras proteins GTPase cycle of Ras proteins GEF GAP GEFs help to dissociate GDP from Ras – active Ras GAPs facilitate GTP hydrolysis – formation of inactive Ras Posttranslational modifications: Interaction with inner part of cytoplasmatic membrane (required for Ras activation) is mediated by posttranslational modifications of CAAX motif at C-terminus of Ras Cys186 of motif CAAX is farnesylated Ras proteins Intermezzo: Prenylation •  Prenylation – the covalent attachment of a lipid consisting of either three (farnesyl) or four (geranylgeranyl) isoprene units (intermediates of cholesterol biosynthesis) to a free thiol of a cysteine side chain at or near the C-terminus of a protein. Prenyl group Intermezzo: Prenylation farnesylation – attachment of farnesyl group (15 C) geranylgeranylation – attachment of geranylgeranyl group (20 C) •  Catalysed by 3 prenyltransferases: farnesyltransferase (FT), geranylgeranyltransferases I and II (GGTI and GGTII). •  There is more than 300 proteins in humans with Cystein at C-terminus, but it is unknown how many are prenylated •  Farnesylation confirmed for: H-, K- and N-Ras, nuclear lamina, CENPE (kinetochors); geranylgeranylation: Rho A and C, Rac1, cdc-42; •  RhoB – farnesylated and geranylgeranylated; •  K-Ras – geranylgeranylovated, if FT is inhibited Ras proteins Posttranslational modifications: Interaction with inner part of cytoplasmatic membrane (required for Ras activation) is mediated by posttranslational modifications of CAAX motif at C-terminus of Ras: •  Cys186 of CAAX motif is farnesylated •  proteolytic cleavage of AAX sequence •  terminal Cys186 is carboxymethylated - protein with these modification is more hydrophobic and has higher affinity to cell membrane For oncogenic activation of Ras is crucial farnesylation! Stucture of Ras proteins •  Heterogenous domain: Ras proteins are highly homologous in first 164AA, the C-terminus (25AA) is variable •  Effector domain (AA 32-40) mediates interaction with effector molecules •  Switch I (30-38) major binding site for GAPs •  Switch II (60-76) binding of GEFs Oncogenic activation of Ras Siqi et al. Nat Rev Cancer, 2018 Dec;18(12):767-777. Oncogenic activation of Ras Oncogenic mutation of Ras detected in almost 35% of human cancer. •  Mutation of H-ras mostly in skin cancer and head and neck squamous carcinoma •  Mutation of K-ras mainly in adenocarcinomas, colorectal carcinomas and pancreatic cancer •  Mutation of N-ras frequent in haematopoetic cancers, mostly in AML and MDS Oncogenic mutations modify Ras activity: •  In codons: 12, 13, 59, 61, 63 – altered proteins have impaired GTPase activity and are resistant to GAPs •  In codons: 16, 19, 116, 117, 119, 144, 146 – resulting proteins have facilitated exchange of GDP for GTP •  Some tumors overexpress Ras Oncogenic activation of Ras 1982: Ras mutation in codon 12 identified (at the same time in 3 laboratories!). It is the first point mutation causally associated with cancer development. Weinberg RA. The Biology of Cancer. Garland Science 2007 Glycine 12 and glutamine 61 are critical for binding of GFP to Ras That is why these codons are mostly mutated in cancer Oncogenic activation of Ras Weinberg RA. The Biology of Cancer. Garland Science 2007 Inhibition of Ras proteins •  for oncogenic features of Ras is crucial farnesylation! Ø  Inhibitors of farnesyl transferases (FTIs) (e.g. CAAX peptides – cell-permeant, specific and efficient, low cytotoxicity) •  By inhibiting farnesylation they prevent post-translational modifications of Ras and thereby its activity •  Potential anti-cancer therapeutics, but: –  Ras proteins have many functions in cells and FTIs blocks all of them (side effects) –  Ras proteins may be also geranylgeranylated (and FTIs do not inhibit geranylgeranyltransferases) –  Ras proteins are not the only targets of FTIs (Rho) FT inhibitors (FTIs) At least 6 inhibitors tested in clinical trials: 1.  BMS-214662 (Bristol-Myers Squibb, Princeton, NJ) 2.  L778123 (Merck and Co., Inc., Whitehouse Station, NJ) 3.  lonafarnib (experimental name SCH66336; Sarasar™; Schering-Plough Corporation, Kenilworth, NJ) 4.  FTI-277 (Calbiochem, EMD Bio-Sciences, San Diego) 5.  L744832 (Biomol International L.P., Plymouth Meeting, PA) 6.  tipifarnib (experimental name R115777; Zarnestra®; Ortho Biotech Products, L.P., Bridgewater, NJ); most advanced testing Lonafarnib: FT inhbitors •  synthetic tricyclic halogenated carboxamide with antineoplastic properties •  used primarily for cancer treatment •  A coctail of lonafarnib and two other drugs proved beneficial for patients with progeria. Tipifarnib: FT inhibitors •  Clinical trials: administration of Tipifarnib to older patients with AML (aged 65 and more) – not approved by FDA, not efficient anti-cancer activity (2005) •  Further tested for efficiency in breast cancer, neurofibromatosis, and other malignacies FT inhibitors (FTIs) •  Not satisfactory outcome in clinical trials so far •  Side effects •  Not clear whether they function primarily via Ras proteins Ras proteins have many effectors Ras triggers 3 key effector pathways 1.  Raf pathway (activation of genes stimulating cell growth/ division, anchorage-independent growth, loss of contact inhibiton) 2.  PI3K pathway (anti-apoptotic effect) 3.  Ral pathway (facilitates invasion and metastatic dissemination of tumors) 1. Ras ⇒ Raf Raf proteins •  Raf: cytosolic proteins, recruited to the cell membrane by activated Ras – and activated itself •  active Raf stimulates MEK (MAP-kinase extracellular signalregulated kinase) by serine phosphorylation •  active MEK is dual kinase: phosphorylates both tyrosine and serine/ threonine of target molecules: including MAP kinases p44 ERK1 a p42 ERK2 (extracellular signal-regulated kinases) •  ERKs have many substrates in cytosol (Rsk, SOS, MEK,...) and (upon nuclear translocation) also nuclear (Ets, Elk-1, SAP-1,...) •  Jun and Fos belong among the target TFs of this pathway •  almost 60-70% melanomas have mutated B-Raf „instead of“ Ras BRAF •  BRAF gene (7q34) has 18 exons, mRNA is 2478 bp long •  Mutations of BRAF are associated with many different cancers: : colorectal carcinoma, malignant melanoma, Non-hodgkins lymfoma, thyroid papillary carcinoma, non-small cell lung carcinoma, lung adenocarcinoma •  80% of mutations is caused by T1799A transversion that leads to amino acid exchange V600E •  This mutation is transforming, because it simulates T599 phosphorylation and/or S602 phosphorylation in activating domain of BRAF; mutated BRAF is thus in permanently active mode, independent on Ras signaling Krajsová I. Dabrafenib v léčbě metastazujícího melanomu. Remedia 24 (3): 215-219, 2013 RASopathy •  RAS/MAPK Syndromes •  Group of neuro-cardio-facio-cutaneous syndromes (NCFC) •  Specific group of developmental disorders caused by germinal mutations of genes encoding proteins of RAS/MAPK signaling cascade •  phenotypically similar group of syndromes: symptoms- e.g. characteristic facial features, cardiac defects, cutaneous abnormalities, neurocognitive delay and a predisposition to malignancies •  Noonan syndrome, neurofibromatosis type 1, Legius syndrome, Costello syndrome a další •  Each syndrome exhibits unique phenotypic features, however, there are numerous overlapping phenotypic features (early diagnosis is difficult) •  Overall RASopathies are one of the largest group of malformation syndromes affecting 1 in 1000 subjects RASopathy •  somatic mutations in RAS/MAPK pathway lead frequently to malignant diseases. That is why much higher rate of cancer development was expected in RASopathies: risk is only 3,5x higher than in normal population; the most prevalent are haematologic malignancies •  Some activating somatic mutations are the same as germinal mutations, but RASopathy-associated mutations are less activating: e.g. The most frequent somatic oncogenic mutation of BRAF (V600E) does not occur in cranio-facio-cutaneous syndrome (typically caused by BRAF mutations) Ø  It is assumed that strongly activating oncogenic mutations are lethal in germ cells Svobodová E. Molekulární diagnostika syndromu Noonanové a přidružených RASopatií pomocí NGS. DP PřF, 2018 Niemeyer CM, Haematologica. 2014 Nov;99(11):1653-62 RASopathies: overview of syndromes Germinal mutations of proto- oncogenes?! Why tolerated?? Ø  Expression limited to a certain tissue (cell type) (RET, c-kit) Ø  Expression of the proto-oncogene occur later during organogenesis thus enabling normal embryogenesis (c-met) Ø  „mildly“ activating mutation (rasopathy, c-kit) Ø  specific types of mutations (cdk 4) 2. Ras ⇒ PI3K PI3K Phosphatidyl inositol 3 – kinase - lipid kinase Ø  Composed of 2 subunits: regulatory p85 and catalytical p110 Ø  catalyze the phosphorylation of the D3 position of the inositol ring of phosphoinositides to produce phosphatidylinositol 3-phosphate (PtdIns3P = PIP3) –secondary messenger triggering downstream signaling PI3K a PIP3 •  PIP3 level is tightly regulated. PIP3 is produced by PI3K and converted back to PIP2 by phosphatases: PTEN, SHIP1, SHIP2 •  PTEN = PIP3 lipid phoshatase; tumor suppressor (breast cancer, glioblastoma, Cowden syndrome) •  PI3K may be activated e.g. by PDGF, NGF, IGF-1, interleukin-3, •  PI3K produces PIP3 and PIP3 interacts with Akt •  Thereby Akt is recruited to the cell membrane, phosphorylated and thus activated Ø  Two mechanisms leading to the Akt hyperactivation : 1.  Aktivation of PI3K 2.  Inactivation of PTEN AKT/PKB •  One of the most common activated protein kinases in cancer •  Hyperactivation of Akt is linked with (1) resistence to apoptosis, (2) enhanced proliferation and (3) motility •  serine/threonine kinases of PKB family (mammalian cells express 3 highly homologous (80%) isoforms of Akt encoded by 3 different genes - AKT1, AKT2 and AKT3 (alias PKBα, PKBβ and PKBγ) Activated AKT ... •  May phoshorylate more than 9000 proteins •  induces antiapoptotic signals, prevents release of cytochromu C from mitochondria, inactivates TF (Forkhead that transactivates FasL), inactivates pro-apoptotic factor Bad and procaspase-9 •  Activates cell cycle: regulates cyclin D stability, blocks transport of p21WAF1 and p27KIP1 into nucleus •  stimulates activity of telomerase via TERT •  induces translocation of MDM2 into nucleus (thereby p53 degradation) •  inactivates GSK3 which leads to increase of β-catenin levels •  inactivates TSC2 (tuberous sclerosis complex 2) that functions as mTOR inhibitor; mTOR is associated with proliferation rate •  AKT often changes subcellular localization = regulates (compartmentalisation) of its substrates Activated AKT ... •  May phoshorylate more than 9000 proteins •  induces antiapoptotic signals, prevents release of cytochromu C from mitochondria, inactivates TF (Forkhead that transactivates FasL), inactivates pro-apoptotic factor Bad and procaspase-9 •  Activates cell cycle: regulates cyclin D stability, blocks transport of p21WAF1 and p27KIP1 into nucleus •  stimulates activity of telomerase via TERT •  induces translocation of MDM2 into nucleus (thereby p53 degradation) •  inactivates GSK3 which leads to increase of β-catenin levels •  inactivates TSC2 (tuberous sclerosis complex 2) that functions as mTOR inhibitor; mTOR is associated with proliferation rate •  AKT often changes subcellular localization = regulates (compartmentalisation) of its substrates Activated AKT ... AKT regulates compartmentalisation of its substrates •  amplification and overexpression of AKT2 in 10-20% ovarian and pancreatic cancer, increased AKT2 activity in 40% of ovarian cancer •  Amplification of AKT1 found in stomach cancers •  Increased activity of AKT1 in prostate and breast cancer – associated with worse prognosis (increased activity in breast carcinomas may be caused by HER2/neu amplification) •  inactivating mutations or deletions of PTEN (prostate and endometrial cancer, glioblastomas, melanomas) •  Amplifications or overexpression of catalytical subunit p110 of PI3K •  activating mutations of ras (around 1/3 of all epithelial tumors) •  Enhanced activity of RTKs, mainly ErbB2/ErbB3 (ErbB3 has docking site for PI3K) AKT in cancerogenesis PI3K pathway alterations in human cancers 3. Ras ⇒ Ral •  Third of the most important Ras effectors: Ral A and Ral B; belong to Ras family, have 58% sequence homology with Ras •  Signaling from Ras to Ral goes through Ral-GEF: activated Ras directly binds to Ral-GEF followed by: a) recruitment of Ral-GEF to the cytosolic side of cell membrane b) conformation change of Ral-GEF → stimulation of GEF activity → activation of Ral-A and Ral-B •  Activated Ral-A and Ral-B activate their target molecules (Rho – actin dynamics) •  Outcome of this activation is increased invasion and metastatic activity of cancer cells 3. Ras ⇒ Ral Ras effector pathways Besides the three described pathways there is more than 8 others Ras effectors. Overall the (Ras) signaling is more complex – rather signaling nets than pathways Thank you for attention!