Jump to content

Hepatocyte growth factor receptor

From Wikipedia, the free encyclopedia

MET
Available structures
PDBOrtholog search:PDBeRCSB
Identifiers
AliasesMET,MET proto-oncogene, receptor tyrosine kinase, AUTS9, HGFR, RCCP2, c-Met, DFNB97, OSFD, c-met
External IDsOMIM:164860;MGI:96969;HomoloGene:206;GeneCards:MET;OMA:MET - orthologs
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_000245
NM_001127500
NM_001324401
NM_001324402

NM_008591

RefSeq (protein)

NP_000236
NP_001120972
NP_001311330
NP_001311331

n/a

Location (UCSC)Chr 7: 116.67 – 116.8 MbChr 6: 17.46 – 17.57 Mb
PubMedsearch[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

Hepatocyte growth factor receptor(HGF receptor)[5][6]is aproteinthat in humans is encoded by theMETgene.The protein possessestyrosine kinaseactivity.[7]The primary single chain precursor protein is post-translationally cleaved to produce the Alpha and beta subunits, which are disulfide linked to form the mature receptor.

HGF receptor is a single pass tyrosine kinase receptor essential for embryonic development, organogenesis and wound healing.Hepatocyte growth factor/Scatter Factor(HGF/SF) and its splicing isoform (NK1, NK2) are the only known ligands of the HGF receptor. MET is normally expressed by cells ofepithelialorigin, while expression of HGF/SF is restricted to cells ofmesenchymalorigin. When HGF/SF binds its cognate receptor MET it induces its dimerization through a not yet completely understood mechanism leading to its activation.

Abnormal MET activation in cancer correlates with poor prognosis, where aberrantly active MET triggers tumor growth, formation of new blood vessels (angiogenesis) that supply the tumor with nutrients, and cancer spread to other organs (metastasis). MET is deregulated in many types of human malignancies, including cancers of kidney, liver, stomach, breast, and brain. Normally, onlystem cellsandprogenitor cellsexpress MET, which allows these cells to grow invasively in order to generate new tissues in an embryo or regenerate damaged tissues in an adult. However,cancer stem cellsare thought to hijack the ability of normal stem cells to express MET, and thus become the cause of cancer persistence and spread to other sites in the body. Both the overexpression of Met/HGFR, as well as itsautocrineactivation by co-expression of its hepatocyte growth factor ligand, have been implicated in oncogenesis.[8][9]

Various mutations in the MET gene are associated withpapillary renal carcinoma.[10]

Gene

[edit]

METproto-oncogene (GeneID: 4233) has a total length of 125,982 bp, and it is located in the 7q31 locus of chromosome 7.[11]METis transcribed into a 6,641 bp mature mRNA, which is then translated into a 1,390 amino-acid MET protein.

Protein

[edit]

MET is areceptor tyrosine kinase(RTK) that is produced as a single-chain precursor. The precursor is proteolytically cleaved at afurinsite to yield a highly glycosylated extracellular α-subunit and a transmembrane β-subunit, which are linked together by adisulfide bridge.[12]

Extracellular

[edit]
  • Region of homology tosemaphorins(Sema domain), which includes the full α-chain and the N-terminal part of the β-chain
  • Cysteine-rich MET-related sequence (MRS domain)
  • Glycine-proline-rich repeats (G-P repeats)
  • Four immunoglobulin-like structures (Ig domains), a typical protein-protein interaction region.[12]

Intracellular

[edit]

A juxtamembrane segment that contains:

  • A serine residue (Ser 985), which inhibits the receptor kinase activity upon phosphorylation[13]
  • A tyrosine residue (Tyr 1003), which is responsible for METpolyubiquitination,endocytosis,and degradation upon interaction with theubiquitin ligaseCBL[14]
  • Tyrosine kinase domain, which mediates MET biological activity. Following MET activation, transphosphorylation occurs on Tyr 1234 and Tyr 1235
  • C-terminal region contains two crucial tyrosines (Tyr 1349 and Tyr 1356), which are inserted into the multisubstrate docking site, capable of recruiting downstream adapter proteins withSrc homology-2(SH2) domains.[15]The two tyrosines of the docking site have been reported to be necessary and sufficient for the signal transduction bothin vitro.[15][16]

MET signaling pathway

[edit]

MET activation by its ligandHGFinduces MET kinase catalytic activity, which triggers transphosphorylation of the tyrosines Tyr 1234 and Tyr 1235. These two tyrosines engage various signal transducers,[17]thus initiating a whole spectrum of biological activities driven by MET, collectively known as the invasive growth program. The transducers interact with the intracellular multisubstrate docking site of MET either directly, such asGRB2,SHC,[18]SRC,and the p85 regulatory subunit of phosphatidylinositol-3 kinase (PI3K),[18]or indirectly through the scaffolding protein Gab1[19]

Tyr 1349 and Tyr 1356 of the multisubstrate docking site are both involved in the interaction with GAB1, SRC, and SHC, while only Tyr 1356 is involved in the recruitment of GRB2, phospholipase C γ (PLC-γ), p85, and SHP2.[20]

GAB1 is a key coordinator of the cellular responses to MET and binds the MET intracellular region with highavidity,but lowaffinity.[21]Upon interaction with MET, GAB1 becomes phosphorylated on several tyrosine residues which, in turn, recruit a number of signalling effectors, includingPI3K,SHP2, and PLC-γ. GAB1 phosphorylation by MET results in a sustained signal that mediates most of the downstream signaling pathways.[22]

Activation of signal transduction

[edit]

MET engagement activates multiplesignal transductionpathways:

Role in development

[edit]

MET mediates a complex program known as invasive growth.[26]Activation of MET triggersmitogenesis,andmorphogenesis.[31][32]

Duringembryonic development,transformation of the flat, two-layergerminal discinto a three-dimensional body depends on transition of some cells from anepithelialphenotypeto spindle-shaped cells with motile behaviour, amesenchymalphenotype.This process is referred to asepithelial-mesenchymal transition(EMT).[33]Later in embryonic development, MET is crucial forgastrulation,angiogenesis,myoblastmigration,bone remodeling,andnervesprouting among others.[34]MET is essential forembryogenesis,becauseMET−/−mice diein uterodue to severe defects in placental development.[35]Along withEctodysplasin A,it has been shown to be involved in the differentiation of anatomical placodes, precursors of scales, feathers and hair follicles in vertebrates.[36]Furthermore, MET is required for such critical processes asliverregeneration andwound healingduring adulthood.[26]

HGF/MET axis is also involved inmyocardial development.Both HGF and MET receptor mRNAs are co-expressed incardiomyocytesfrom E7.5, soon after the heart has been determined, to E9.5. Transcripts for HGF ligand and receptor are first detected before the occurrence of cardiac beating and looping, and persist throughout the looping stage, when heart morphology begins to elaborate.[37]In avian studies, HGF was found in the myocardial layer of the atrioventricular canal, in a developmental stage in which the epithelial to mesenchymal transformation (EMT) of theendocardial cushionoccurs.[38]However, MET is not essential for heart development, since α-MHCMet-KO mice show normal heart development.[39]

Expression

[edit]

Tissue distribution

[edit]

MET is normally expressed byepithelial cells.[26]However, MET is also found onendothelial cells,neurons,hepatocytes,hematopoieticcells,melanocytesand neonatal cardiomyocytes.[32][40]HGF expression is restricted to cells ofmesenchymalorigin.[33]

Transcriptional control

[edit]

METtranscription is activated by HGF and severalgrowth factors.[41]METpromoterhas four putative binding sites forEts,a family oftranscription factorsthat control several invasive growth genes.[41]ETS1activatesMETtranscriptionin vitro.[42]METtranscription is activated byhypoxia-inducible factor 1(HIF1), which is activated by low concentration of intracellular oxygen.[43]HIF1 can bind to one of the severalhypoxiaresponse elements(HREs) in theMETpromoter.[33]Hypoxia also activates transcription factorAP-1,which is involved inMETtranscription.[33]

Clinical significance

[edit]

Role in cancer

[edit]

MET pathway plays an important role in the development of cancer through:

Coordinated down-regulation of both MET and its downstream effector extracellular signal-regulated kinase 2 (ERK2) bymiR-199a*may be effective in inhibiting not only cell proliferation but also motility and invasive capabilities of tumor cells.[45]

MET amplification has emerged as a potential biomarker of theclear cell tumorsubtype.[46]

The amplification of thecell surface receptorMET often drives resistance toanti-EGFR therapiesincolorectal cancer.[47]

Role in autism

[edit]

The SFARIgene database lists MET with anautismscore of 2.0, which indicates that it is a strong candidate for playing a role in cases of autism. The database also identifies at least one study that found a role for MET in cases ofschizophrenia.The gene was first implicated in autism in a study that identified a polymorphism in the promoter of the MET gene.[48]The polymorphism reduces transcription by 50%. Further, the variant as an autism risk polymorphism has been replicated, and shown to be enriched in children with autism and gastrointestinal disturbances.[49]A rare mutation has been found that appears in two family members, one with autism and the other with a social and communication disorder.[50]The role of the receptor in brain development is distinct from its role in other developmental processes. Activation of the MET receptor regulates synapse formation[51][52][53][54][55]and can impact the development and function of circuits involved in social and emotional behavior.[56]

Role in heart function

[edit]

In adult mice, MET is required to protect cardiomyocytes by preventing age-relatedoxidative stress,apoptosis,fibrosis and cardiac dysfunction.[39]Moreover, MET inhibitors, such ascrizotinibor PF-04254644, have been tested by short-term treatments in cellular and preclinical models, and have been shown to induce cardiomyocytes death through ROS production, activation ofcaspases,metabolism alteration and blockage ofion channels.[57][58]

In the injured heart, HGF/MET axis plays important roles in cardioprotection by promoting pro-survival (anti-apoptotic and anti-autophagic) effects in cardiomyocytes, angiogenesis, inhibition of fibrosis, anti-inflammatory and immunomodulatory signals, and regeneration through activation ofcardiac stem cells.[59][60]

Interaction with tumour suppressor genes

[edit]

PTEN

[edit]

PTEN(phosphatase and tensin homolog) is atumor suppressor geneencoding a protein PTEN, which possesses lipid and protein phosphatase-dependent as well as phosphatase-independent activities.[61]PTEN proteinphosphataseis able to interfere with MET signaling by dephosphorylating either PIP3generated byPI3K,or the p52 isoform ofSHC.SHC dephosphorylation inhibits recruitment of theGRB2adapter to activated MET.[29]

VHL

[edit]

There is evidence of correlation between inactivation ofVHLtumor suppressor geneand increased MET signaling inrenal cell carcinoma(RCC) and also in malignant transformations of the heart.[62][63]

Cancer therapies targeting HGF/MET

[edit]

Since tumor invasion and metastasis are the main cause of death in cancer patients, interfering with MET signaling appears to be a promising therapeutic approach. A comprehensive list of HGF and MET targeted experimental therapeutics for oncology now in human clinical trials can be foundhere.

MET kinase inhibitors

[edit]

Kinase inhibitors are low molecular weight molecules that preventATPbinding to MET, thus inhibiting receptor transphosphorylation and recruitment of the downstream effectors. The limitations of kinase inhibitors include the facts that they only inhibit kinase-dependent MET activation, and that none of them is fully specific for MET.

  • K252a(FermentekBiotechnology) is astaurosporineanalogue isolated fromNocardiopsissp. soilfungi,and it is a potent inhibitor of allreceptor tyrosine kinases(RTKs). At nanomolar concentrations, K252a inhibits both thewild typeand the mutant (M1268T) MET function.[64]
  • SU11274 (SUGEN) specifically inhibits MET kinase activity and its subsequent signaling. SU11274 is also an effective inhibitor of the M1268T and H1112Y MET mutants, but not the L1213V and Y1248H mutants.[65]SU11274 has been demonstrated to inhibit HGF-induced motility and invasion of epithelial and carcinoma cells.[66]
  • PHA-665752 (Pfizer) specifically inhibits MET kinase activity, and it has been demonstrated to represses both HGF-dependent and constitutive MET phosphorylation.[67]Furthermore, some tumors harboringMETamplifications are highly sensitive to treatment with PHA-665752.[68]
  • Tivantinib(ArQule) is a promising selective inhibitor of MET, which entered a phase 2 clinical trial in 2008. (Failed a phase 3 in 2017)
  • Foretinib(XL880, Exelixis) targets multiplereceptor tyrosine kinases(RTKs) with growth-promoting and angiogenic properties. The primary targets of foretinib are MET,VEGFR2,andKDR.Foretinib has completed a phase 2 clinical trials with indications for papillaryrenal cell carcinoma,gastric cancer,andhead and neck cancer[citation needed]
  • SGX523 (SGX Pharmaceuticals) specifically inhibits MET at low nanomolar concentrations.
  • MP470 (SuperGen) is a novel inhibitor ofc-KIT,MET,PDGFR,Flt3, andAXL.Phase I clinical trial of MP470 had been announced in 2007.
  • Vebreltinib,approved in China for the treatment of non-small-cell lung cancer.[69]

HGF inhibitors

[edit]

Since HGF is the only known ligand of MET, blocking the formation of a HGF:MET complex blocks METbiological activity.For this purpose, truncated HGF, anti-HGF neutralizing antibodies, and an uncleavable form of HGF have been utilized so far. The major limitation of HGF inhibitors is that they block only HGF-dependent MET activation.

  • NK4 competes with HGF as it binds MET without inducing receptor activation, thus behaving as a fullantagonist.NK4 is a molecule bearing the N-terminal hairpin and the fourkringle domainsof HGF. Moreover, NK4 is structurally similar toangiostatins,which is why it possesses anti-angiogenic activity.[70]
  • Neutralizing anti-HGF antibodies were initially tested in combination, and it was shown that at least threeantibodies,acting on different HGFepitopes,are necessary to prevent MET tyrosine kinase activation.[71]More recently, it has been demonstrated that fully humanmonoclonal antibodiescan individually bind and neutralize human HGF, leading to regression of tumors in mouse models.[72]Two anti-HGF antibodies are currently available: the humanized AV299 (AVEO), and the fully human AMG102 (Amgen).
  • Uncleavable HGF is an engineered form of pro-HGF carrying a single amino-acid substitution, which prevents the maturation of the molecule. Uncleavable HGF is capable of blocking MET-induced biological responses by binding MET with high affinity and displacing mature HGF. Moreover, uncleavable HGF competes with the wild-type endogenous pro-HGF for the catalytic domain ofproteasesthat cleave HGF precursors. Local and systemic expression of uncleavable HGF inhibits tumor growth and, more importantly, preventsmetastasis.

Decoy MET

[edit]

Decoy MET refers to a soluble truncated MET receptor. Decoys are able to inhibit MET activation mediated by both HGF-dependent and independent mechanisms, as decoys prevent both the ligand binding and the MET receptor homodimerization. CGEN241 (Compugen) is a decoy MET that is highly efficient in inhibiting tumor growth and preventingmetastasisin animal models.[73]

Immunotherapy targeting MET

[edit]

Drugs used forimmunotherapycan act either passively by enhancing the immunologic response to MET-expressing tumor cells, or actively by stimulatingimmune cellsand altering differentiation/growth of tumor cells.[74]

Passive immunotherapy

[edit]

Administeringmonoclonal antibodies(mAbs) is a form of passive immunotherapy. MAbs facilitate destruction of tumor cells bycomplement-dependent cytotoxicity(CDC) and cell-mediated cytotoxicity (ADCC). In CDC, mAbs bind to specificantigen,leading to activation of thecomplement cascade,which in turn leads to formation of pores in tumor cells. In ADCC, the Fab domain of a mAb binds to atumor antigen,and Fc domain binds to Fc receptors present on effector cells (phagocytesandNK cells), thus forming a bridge between an effector and a target cells. This induces the effector cell activation, leading tophagocytosisof the tumor cell byneutrophilsandmacrophages.Furthermore,NK cellsreleasecytotoxicmolecules, which lyse tumor cells.[74]

  • DN30 is monoclonal anti-MET antibody that recognizes the extracellular portion of MET. DN30 induces bothsheddingof the METectodomainas well as cleavage of the intracellular domain, which is successively degraded byproteasomemachinery. As a consequence, on one side MET is inactivated, and on the other side the shed portion of extracellular MET hampers activation of other MET receptors, acting as a decoy. DN30 inhibits tumour growth and preventsmetastasisin animal models.[75]
  • OA-5D5 is one-armed monoclonal anti-MET antibody that was demonstrated to inhibit orthotopic pancreatic[76]andglioblastoma[77]tumor growth and to improve survival intumor xenograft models.OA-5D5 is produced as arecombinant proteininEscherichia coli.It is composed of murinevariable domainsfor theheavyandlight chainswith human IgG1 constant domains. The antibody blocks HGF binding to MET in a competitive fashion.

Active immunotherapy

[edit]

Active immunotherapyto MET-expressing tumors can be achieved by administeringcytokines,such asinterferons(IFNs) andinterleukins(IL-2), which triggers non-specific stimulation of numerous immune cells. IFNs have been tested as therapies for many types of cancers and have demonstrated therapeutic benefits. IL-2 has been approved by theU.S. Food and Drug Administration(FDA) for the treatment ofrenal cell carcinomaand metastatic melanoma, which often have deregulated MET activity.[74]

Interactions

[edit]

Met has been shown tointeractwith:

See also

[edit]

References

[edit]
  1. ^abcGRCh38: Ensembl release 89: ENSG00000105976Ensembl,May 2017
  2. ^abcGRCm38: Ensembl release 89: ENSMUSG00000009376Ensembl,May 2017
  3. ^"Human PubMed Reference:".National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^"Mouse PubMed Reference:".National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF,Aaronson SA(February 1991). "Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product".Science.251(4995): 802–4.Bibcode:1991Sci...251..802B.doi:10.1126/science.1846706.PMID1846706.
  6. ^Galland F, Stefanova M, Lafage M, Birnbaum D (1992). "Localization of the 5' end of the MCF2 oncogene to human chromosome 15q15----q23".Cytogenet. Cell Genet.60(2): 114–6.doi:10.1159/000133316.PMID1611909.
  7. ^Cooper CS (January 1992). "The met oncogene: from detection by transfection to transmembrane receptor for hepatocyte growth factor".Oncogene.7(1): 3–7.PMID1531516.
  8. ^Johnson M, Koukoulis G, Kochhar K, Kubo C, Nakamura T, Iyer A (September 1995). "Selective tumorigenesis in non-parenchymal liver epithelial cell lines by hepatocyte growth factor transfection".Cancer Letters.96(1): 37–48.doi:10.1016/0304-3835(95)03915-j.PMID7553606.
  9. ^Kochhar KS, Johnson ME, Volpert O, Iyer AP (1995). "Evidence for autocrine basis of transformation in NIH-3T3 cells transfected with met/HGF receptor gene".Growth Factors.12(4): 303–13.doi:10.3109/08977199509028968.PMID8930021.
  10. ^"Entrez Gene: MET met proto-oncogene (hepatocyte growth factor receptor)".
  11. ^Dean M, Park M, Le Beau MM, Robins TS, Diaz MO, Rowley JD, Blair DG, Vande Woude GF (1985)."The human met oncogene is related to the tyrosine kinase oncogenes".Nature.318(6044): 385–8.Bibcode:1985Natur.318..385D.doi:10.1038/318385a0.PMID4069211.S2CID4359961.
  12. ^abBirchmeier C, Birchmeier W, Gherardi E, Vande Woude GF (December 2003). "Met, metastasis, motility and more".Nat. Rev. Mol. Cell Biol.4(12): 915–25.doi:10.1038/nrm1261.PMID14685170.S2CID19330786.
  13. ^Gandino L, Longati P, Medico E, Prat M, Comoglio PM (January 1994)."Phosphorylation of serine 985 negatively regulates the hepatocyte growth factor receptor kinase".J. Biol. Chem.269(3): 1815–20.doi:10.1016/S0021-9258(17)42099-0.PMID8294430.
  14. ^Peschard P, Fournier TM, Lamorte L, Naujokas MA, Band H, Langdon WY, Park M (November 2001)."Mutation of the c-Cbl TKB domain binding site on the Met receptor tyrosine kinase converts it into a transforming protein".Mol. Cell.8(5): 995–1004.doi:10.1016/S1097-2765(01)00378-1.PMID11741535.
  15. ^abPonzetto C, Bardelli A, Zhen Z, Maina F, dalla Zonca P, Giordano S, Graziani A, Panayotou G, Comoglio PM (April 1994). "A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family".Cell.77(2): 261–71.doi:10.1016/0092-8674(94)90318-2.PMID7513258.S2CID23383203.
  16. ^Maina F, Casagranda F, Audero E, Simeone A, Comoglio PM, Klein R, Ponzetto C (November 1996)."Uncoupling of Grb2 from the Met receptor in vivo reveals complex roles in muscle development".Cell.87(3): 531–42.doi:10.1016/S0092-8674(00)81372-0.PMID8898205.S2CID12943699.
  17. ^Johnson M, Kochhar K, Nakamura T, Iyer A (July 1995). "Hepatocyte growth factor-induced signal transduction in two normal mouse epithelial cell lines".Biochemistry and Molecular Biology International.36(3): 465–74.PMID7549943.
  18. ^abPelicci G, Giordano S, Zhen Z, Salcini AE, Lanfrancone L, Bardelli A, Panayotou G, Waterfield MD, Ponzetto C, Pelicci PG (April 1995). "The motogenic and mitogenic responses to HGF are amplified by the Shc adaptor protein".Oncogene.10(8): 1631–8.PMID7731718.
  19. ^Weidner KM, Di Cesare S, Sachs M, Brinkmann V, Behrens J, Birchmeier W (November 1996). "Interaction between Gab1 and the c-Met receptor tyrosine kinase is responsible for epithelial morphogenesis".Nature.384(6605): 173–6.Bibcode:1996Natur.384..173W.doi:10.1038/384173a0.PMID8906793.S2CID4357372.
  20. ^Furge KA, Zhang YW, Vande Woude GF (November 2000). "Met receptor tyrosine kinase: enhanced signaling through adapter proteins".Oncogene.19(49): 5582–9.doi:10.1038/sj.onc.1203859.PMID11114738.S2CID22385297.
  21. ^Gual P, Giordano S, Anguissola S, Parker PJ, Comoglio PM (January 2001). "Gab1 phosphorylation: a novel mechanism for negative regulation of HGF receptor signaling".Oncogene.20(2): 156–66.doi:10.1038/sj.onc.1204047.PMID11313945.S2CID35447713.
  22. ^Gual P, Giordano S, Williams TA, Rocchi S, Van Obberghen E, Comoglio PM (March 2000). "Sustained recruitment of phospholipase C-gamma to Gab1 is required for HGF-induced branching tubulogenesis".Oncogene.19(12): 1509–18.doi:10.1038/sj.onc.1203514.PMID10734310.S2CID22727382.
  23. ^O'Brien LE, Tang K, Kats ES, Schutz-Geschwender A, Lipschutz JH,Mostov KE(July 2004)."ERK and MMPs sequentially regulate distinct stages of epithelial tubule development".Dev. Cell.7(1): 21–32.doi:10.1016/j.devcel.2004.06.001.PMID15239951.
  24. ^Marshall CJ (January 1995)."Specificity of receptor tyrosine kinase signaling: transient versus sustained extracellular signal-regulated kinase activation".Cell.80(2): 179–85.doi:10.1016/0092-8674(95)90401-8.PMID7834738.S2CID8995643.
  25. ^Graziani A, Gramaglia D, Cantley LC, Comoglio PM (November 1991)."The tyrosine-phosphorylated hepatocyte growth factor/scatter factor receptor associates with phosphatidylinositol 3-kinase".J. Biol. Chem.266(33): 22087–90.doi:10.1016/S0021-9258(18)54536-1.PMID1718989.
  26. ^abcdGentile A, Trusolino L, Comoglio PM (March 2008). "The Met tyrosine kinase receptor in development and cancer".Cancer Metastasis Rev.27(1): 85–94.doi:10.1007/s10555-007-9107-6.PMID18175071.S2CID33076010.
  27. ^Boccaccio C, Andò M, Tamagnone L, Bardelli A, Michieli P, Battistini C, Comoglio PM (January 1998). "Induction of epithelial tubules by growth factor HGF depends on the STAT pathway".Nature.391(6664): 285–8.Bibcode:1998Natur.391..285B.doi:10.1038/34657.PMID9440692.S2CID30330705.
  28. ^Monga SP, Mars WM, Pediaditakis P, Bell A, Mulé K, Bowen WC, Wang X, Zarnegar R, Michalopoulos GK (April 2002). "Hepatocyte growth factor induces Wnt-independent nuclear translocation of beta-catenin after Met-beta-catenin dissociation in hepatocytes".Cancer Res.62(7): 2064–71.PMID11929826.
  29. ^abAbounader R, Reznik T, Colantuoni C, Martinez-Murillo F, Rosen EM, Laterra J (December 2004). "Regulation of c-Met-dependent gene expression by PTEN".Oncogene.23(57): 9173–82.doi:10.1038/sj.onc.1208146.PMID15516982.S2CID7623249.
  30. ^Gude NA, Emmanuel G, Wu W, Cottage CT, Fischer K, Quijada P, Muraski JA, Alvarez R, Rubio M, Schaefer E, Sussman MA (May 2008)."Activation of Notch-mediated protective signaling in the myocardium".Circ. Res.102(9): 1025–35.doi:10.1161/CIRCRESAHA.107.164749.PMC3760732.PMID18369158.
  31. ^Johnson M, Koukoulis G, Matsumoto K, Nakamura T, Iyer A (June 1993)."Hepatocyte growth factor induces proliferation and morphogenesis in nonparenchymal epithelial liver cells".Hepatology.17(6): 1052–61.doi:10.1016/0270-9139(93)90122-4.PMID8514254.
  32. ^ab"he fields of HGF/c-Met involvement".HealthValue. Archived fromthe originalon 27 September 2007.Retrieved13 June2009.
  33. ^abcdBoccaccio C, Comoglio PM (August 2006). "Invasive growth: a MET-driven genetic programme for cancer and stem cells".Nat. Rev. Cancer.6(8): 637–45.doi:10.1038/nrc1912.PMID16862193.S2CID396385.
  34. ^Birchmeier C, Gherardi E (October 1998). "Developmental roles of HGF/SF and its receptor, the c-Met tyrosine kinase".Trends Cell Biol.8(10): 404–10.doi:10.1016/S0962-8924(98)01359-2.PMID9789329.
  35. ^Uehara Y, Minowa O, Mori C, Shiota K, Kuno J, Noda T, Kitamura N (February 1995). "Placental defect and embryonic lethality in mice lacking hepatocyte growth factor/scatter factor".Nature.373(6516): 702–5.Bibcode:1995Natur.373..702U.doi:10.1038/373702a0.PMID7854453.S2CID4361262.
  36. ^Barrow-McGee R, Kishi N, Joffre C, Ménard L, Hervieu A, Bakhouche BA, et al. (2016)."Beta 1-integrin-c-Met cooperation reveals an inside-in survival signalling on autophagy-related endomembranes".Nature Communications.7:11942.Bibcode:2016NatCo...711942B.doi:10.1038/ncomms11942.PMC4931016.PMID27336951.
  37. ^Rappolee DA, Iyer A, Patel Y (June 1996)."Hepatocyte growth factor and its receptor are expressed in cardiac myocytes during early cardiogenesis".Circulation Research.78(6): 1028–36.doi:10.1161/01.RES.78.6.1028.PMID8635233.
  38. ^Song W, Majka SM, McGuire PG (1999)."Hepatocyte growth factor expression in the developing myocardium: evidence for a role in the regulation of the mesenchymal cell phenotype and urokinase expression".Developmental Dynamics.214(1): 92–100.doi:10.1002/(SICI)1097-0177(199901)214:1<92::AID-DVDY9>3.0.CO;2-X.PMID9915579.
  39. ^abArechederra M, Carmona R, González-Nuñez M, Gutiérrez-Uzquiza A, Bragado P, Cruz-González I, Cano E, Guerrero C, Sánchez A, López-Novoa JM, Schneider MD, Maina F, Muñoz-Chápuli R, Porras A (December 2013)."Met signaling in cardiomyocytes is required for normal cardiac function in adult mice"(PDF).Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease.1832(12): 2204–15.doi:10.1016/j.bbadis.2013.08.008.PMID23994610.
  40. ^Leo C, Sala V, Morello M, Chiribiri A, Riess I, Mancardi D, Schiaffino S, Ponzetto C, Crepaldi T (9 February 2011)."Activated Met signalling in the developing mouse heart leads to cardiac disease".PLOS ONE.6(2): e14675.Bibcode:2011PLoSO...614675L.doi:10.1371/journal.pone.0014675.PMC3036588.PMID21347410.
  41. ^abShirasaki F, Makhluf HA, LeRoy C, Watson DK, Trojanowska M (December 1999)."Ets transcription factors cooperate with Sp1 to activate the human tenascin-C promoter".Oncogene.18(54): 7755–64.doi:10.1038/sj.onc.1203360.PMID10618716.
  42. ^Gambarotta G, Boccaccio C, Giordano S, Andŏ M, Stella MC, Comoglio PM (November 1996). "Ets up-regulates MET transcription".Oncogene.13(9): 1911–7.PMID8934537.
  43. ^Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM (April 2003)."Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene".Cancer Cell.3(4): 347–61.doi:10.1016/S1535-6108(03)00085-0.PMID12726861.
  44. ^"HGF/c-Met and cancer".HealthValue. Archived fromthe originalon 27 September 2007.Retrieved13 June2009.
  45. ^Kim S, Lee UJ, Kim MN, Lee EJ, Kim JY, Lee MY, Choung S, Kim YJ, Choi YC (June 2008)."MicroRNA miR-199a* regulates the MET proto-oncogene and the downstream extracellular signal-regulated kinase 2 (ERK2)".J. Biol. Chem.283(26): 18158–66.doi:10.1074/jbc.M800186200.PMID18456660.
  46. ^del Carmen MG, Birrer M, Schorge JO (September 2012). "Clear cell carcinoma of the ovary: a review of the literature".Gynecol. Oncol.126(3): 481–90.doi:10.1016/j.ygyno.2012.04.021.PMID22525820.
  47. ^Bardelli A,Corso S, Bertotti A, Hobor S, Valtorta E, Siravegna G, Sartore-Bianchi A, Scala E, Cassingena A, Zecchin D, Apicella M, Migliardi G, Galimi F, Lauricella C, Zanon C, Perera T, Veronese S, Corti G, Amatu A, Gambacorta M, Diaz LA, Sausen M, Velculescu VE, Comoglio P, Trusolino L, Di Nicolantonio F, Giordano S, Siena S (June 2013)."Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer".Cancer Discov.3(6): 658–73.doi:10.1158/2159-8290.CD-12-0558.PMC4078408.PMID23729478.
  48. ^Campbell DB, Sutcliffe JS, Ebert PJ, Militerni R, Bravaccio C, Trillo S, Elia M, Schneider C, Melmed R, Sacco R, Persico AM, Levitt P (2006)."A genetic variant that disrupts MET transcription is associated with autism".Proc. Natl. Acad. Sci. U.S.A.103(45): 16834–9.doi:10.1073/pnas.0605296103.PMC1838551.PMID17053076.
  49. ^Campbell DB, Buie TM, Winter H, Bauman M, Sutcliffe JS, Perrin JM, Levitt P (2009). "Distinct genetic risk based on association of MET in families with co-occurring autism and gastrointestinal conditions".Pediatrics.123(3): 1018–24.doi:10.1542/peds.2008-0819.PMID19255034.S2CID5395283.
  50. ^Lambert N, Wermenbol V, Pichon B, Acosta S, van den Ameele J, Perazzolo C, Messina D, Musumeci MF, Dessars B, De Leener A, Abramowicz M, Vilain C (2014). "A familial heterozygous null mutation of MET in autism spectrum disorder".Autism Res.7(5): 617–22.doi:10.1002/aur.1396.PMID24909855.S2CID5608613.
  51. ^Qiu S, Lu Z, Levitt P (2014)."MET receptor tyrosine kinase controls dendritic complexity, spine morphogenesis, and glutamatergic synapse maturation in the hippocampus".J. Neurosci.34(49): 16166–79.doi:10.1523/JNEUROSCI.2580-14.2014.PMC4252539.PMID25471559.
  52. ^Eagleson KL, Milner TA, Xie Z, Levitt P (2013)."Synaptic and extrasynaptic location of the receptor tyrosine kinase met during postnatal development in the mouse neocortex and hippocampus".J. Comp. Neurol.521(14): 3241–59.doi:10.1002/cne.23343.PMC3942873.PMID23787772.
  53. ^Judson MC, Eagleson KL, Levitt P (2011)."A new synaptic player leading to autism risk: Met receptor tyrosine kinase".J Neurodev Disord.3(3): 282–92.doi:10.1007/s11689-011-9081-8.PMC3261279.PMID21509596.
  54. ^Qiu S, Anderson CT, Levitt P, Shepherd GM (2011)."Circuit-specific intracortical hyperconnectivity in mice with deletion of the autism-associated Met receptor tyrosine kinase".J. Neurosci.31(15): 5855–64.doi:10.1523/JNEUROSCI.6569-10.2011.PMC3086026.PMID21490227.
  55. ^Judson MC, Eagleson KL, Wang L, Levitt P (2010)."Evidence of cell-nonautonomous changes in dendrite and dendritic spine morphology in the met-signaling-deficient mouse forebrain".J. Comp. Neurol.518(21): 4463–78.doi:10.1002/cne.22467.PMC2952412.PMID20853516.
  56. ^Rudie JD, Hernandez LM, Brown JA, Beck-Pancer D, Colich NL, Gorrindo P, Thompson PM, Geschwind DH, Bookheimer SY, Levitt P, Dapretto M (2012)."Autism-associated promoter variant in MET impacts functional and structural brain networks".Neuron.75(5): 904–15.doi:10.1016/j.neuron.2012.07.010.PMC3454529.PMID22958829.
  57. ^Doherty KR, Wappel RL, Talbert DR, Trusk PB, Moran DM, Kramer JW, Brown AM, Shell SA, Bacus S (October 2013). "Multi-parameter in vitro toxicity testing of crizotinib, sunitinib, erlotinib, and nilotinib in human cardiomyocytes".Toxicology and Applied Pharmacology.272(1): 245–55.doi:10.1016/j.taap.2013.04.027.PMID23707608.
  58. ^Aguirre SA, Heyen JR, Collette W, Bobrowski W, Blasi ER (April 2010)."Cardiovascular effects in rats following exposure to a receptor tyrosine kinase inhibitor".Toxicologic Pathology.38(3): 416–28.doi:10.1177/0192623310364027.PMID20231546.
  59. ^Schmoldt A, Benthe HF, Haberland G, Scott WA, Mahoney E, Pounds JG, Long GJ, Rosen JF (February 1991)."Cellular and molecular toxicity of lead in bone".Environmental Health Perspectives.91(17): 17–32.doi:10.1289/ehp.919117.PMC1519349.PMID2040247.
  60. ^Sala V, Crepaldi T (May 2011)."Novel therapy for myocardial infarction: can HGF/Met be beneficial?".Cellular and Molecular Life Sciences.68(10): 1703–17.doi:10.1007/s00018-011-0633-6.PMC11114731.PMID21327916.S2CID32535928.
  61. ^Maehama T, Dixon JE (May 1998)."The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate".J. Biol. Chem.273(22): 13375–8.doi:10.1074/jbc.273.22.13375.PMID9593664.
  62. ^Morris MR, Gentle D, Abdulrahman M, Maina EN, Gupta K, Banks RE, Wiesener MS, Kishida T, Yao M, Teh B, Latif F, Maher ER (June 2005)."Tumor suppressor activity and epigenetic inactivation of hepatocyte growth factor activator inhibitor type 2/SPINT2 in papillary and clear cell renal cell carcinoma".Cancer Res.65(11): 4598–606.doi:10.1158/0008-5472.CAN-04-3371.PMID15930277.
  63. ^Lei L, Mason S, Liu D, Huang Y, Marks C, Hickey R, Jovin IS, Pypaert M, Johnson RS, Giordano FJ (June 2008)."Hypoxia-inducible factor-dependent degeneration, failure, and malignant transformation of the heart in the absence of the von Hippel-Lindau protein".Molecular and Cellular Biology.28(11): 3790–803.doi:10.1128/MCB.01580-07.PMC2423296.PMID18285456.
  64. ^Morotti A, Mila S, Accornero P, Tagliabue E, Ponzetto C (July 2002). "K252a inhibits the oncogenic properties of Met, the HGF receptor".Oncogene.21(32): 4885–93.doi:10.1038/sj.onc.1205622.PMID12118367.S2CID32305287.
  65. ^Berthou S, Aebersold DM, Schmidt LS, Stroka D, Heigl C, Streit B, Stalder D, Gruber G, Liang C, Howlett AR, Candinas D, Greiner RH, Lipson KE, Zimmer Y (July 2004). "The Met kinase inhibitor SU11274 exhibits a selective inhibition pattern toward different receptor mutated variants".Oncogene.23(31): 5387–93.doi:10.1038/sj.onc.1207691.PMID15064724.S2CID12545344.
  66. ^Wang X, Le P, Liang C, Chan J, Kiewlich D, Miller T, Harris D, Sun L, Rice A, Vasile S, Blake RA, Howlett AR, Patel N, McMahon G, Lipson KE (November 2003). "Potent and selective inhibitors of the Met [hepatocyte growth factor/scatter factor (HGF/SF) receptor] tyrosine kinase block HGF/SF-induced tumor cell growth and invasion".Mol. Cancer Ther.2(11): 1085–92.PMID14617781.
  67. ^Christensen JG, Schreck R, Burrows J, Kuruganti P, Chan E, Le P, Chen J, Wang X, Ruslim L, Blake R, Lipson KE, Ramphal J, Do S, Cui JJ, Cherrington JM, Mendel DB (November 2003). "A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo".Cancer Res.63(21): 7345–55.PMID14612533.
  68. ^Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, Kim WJ, Okimoto RA, Bell DW, Sgroi DC, Christensen JG, Settleman J, Haber DA (February 2006)."Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752".Proc. Natl. Acad. Sci. U.S.A.103(7): 2316–21.Bibcode:2006PNAS..103.2316S.doi:10.1073/pnas.0508776103.PMC1413705.PMID16461907.
  69. ^"Vebreltinib Receives Approval in China For MET Exon 14+ NSCLC".onclive.16 November 2023.
  70. ^Matsumoto K, Nakamura T (April 2003)."NK4 (HGF-antagonist/angiogenesis inhibitor) in cancer biology and therapeutics".Cancer Sci.94(4): 321–7.doi:10.1111/j.1349-7006.2003.tb01440.x.PMC11160298.PMID12824898.S2CID24806218.
  71. ^Cao B, Su Y, Oskarsson M, Zhao P, Kort EJ, Fisher RJ, Wang LM, Vande Woude GF (June 2001)."Neutralizing monoclonal antibodies to hepatocyte growth factor/scatter factor (HGF/SF) display antitumor activity in animal models".Proc. Natl. Acad. Sci. U.S.A.98(13): 7443–8.Bibcode:2001PNAS...98.7443C.doi:10.1073/pnas.131200498.PMC34688.PMID11416216.
  72. ^Burgess T, Coxon A, Meyer S, Sun J, Rex K, Tsuruda T, Chen Q, Ho SY, Li L, Kaufman S, McDorman K, Cattley RC, Sun J, Elliott G, Zhang K, Feng X, Jia XC, Green L, Radinsky R, Kendall R (February 2006). "Fully human monoclonal antibodies to hepatocyte growth factor with therapeutic potential against hepatocyte growth factor/c-Met-dependent human tumors".Cancer Res.66(3): 1721–9.doi:10.1158/0008-5472.CAN-05-3329.PMID16452232.
  73. ^Michieli P, Mazzone M, Basilico C, Cavassa S, Sottile A, Naldini L, Comoglio PM (July 2004)."Targeting the tumor and its microenvironment by a dual-function decoy Met receptor".Cancer Cell.6(1): 61–73.doi:10.1016/j.ccr.2004.05.032.PMID15261142.
  74. ^abcReang P, Gupta M, Kohli K (2006)."Biological response modifiers in cancer".MedGenMed.8(4): 33.PMC1868326.PMID17415315.
  75. ^Petrelli A, Circosta P, Granziero L, Mazzone M, Pisacane A, Fenoglio S, Comoglio PM, Giordano S (March 2006)."Ab-induced ectodomain shedding mediates hepatocyte growth factor receptor down-regulation and hampers biological activity".Proc. Natl. Acad. Sci. U.S.A.103(13): 5090–5.Bibcode:2006PNAS..103.5090P.doi:10.1073/pnas.0508156103.PMC1458799.PMID16547140.
  76. ^Jin H, Yang R, Zheng Z, Romero M, Ross J, Bou-Reslan H, Carano RA, Kasman I, Mai E, Young J, Zha J, Zhang Z, Ross S, Schwall R, Colbern G, Merchant M (June 2008)."MetMAb, the one-armed 5D5 anti-c-Met antibody, inhibits orthotopic pancreatic tumor growth and improves survival".Cancer Res.68(11): 4360–8.doi:10.1158/0008-5472.CAN-07-5960.PMID18519697.
  77. ^Martens T, Schmidt NO, Eckerich C, Fillbrandt R, Merchant M, Schwall R, Westphal M, Lamszus K (October 2006)."A novel one-armed anti-c-Met antibody inhibits glioblastoma growth in vivo".Clin. Cancer Res.12(20 Pt 1): 6144–52.doi:10.1158/1078-0432.CCR-05-1418.PMID17062691.
  78. ^Davies G, Jiang WG, Mason MD (2001). "HGF/SF modifies the interaction between its receptor c-Met, and the E-cadherin/catenin complex in prostate cancer cells".Int. J. Mol. Med.7(4): 385–8.doi:10.3892/ijmm.7.4.385.PMID11254878.
  79. ^Petrelli A, Gilestro GF, Lanzardo S, Comoglio PM, Migone N, Giordano S (2002). "The endophilin-CIN85-Cbl complex mediates ligand-dependent downregulation of c-Met".Nature.416(6877): 187–90.Bibcode:2002Natur.416..187P.doi:10.1038/416187a.PMID11894096.S2CID4389099.
  80. ^Ng C, Jackson RA, Buschdorf JP, Sun Q, Guy GR, Sivaraman J (2008)."Structural basis for a novel intrapeptidyl H-bond and reverse binding of c-Cbl-TKB domain substrates".EMBO J.27(5): 804–16.doi:10.1038/emboj.2008.18.PMC2265755.PMID18273061.
  81. ^Grisendi S, Chambraud B, Gout I, Comoglio PM, Crepaldi T (2001)."Ligand-regulated binding of FAP68 to the hepatocyte growth factor receptor".J. Biol. Chem.276(49): 46632–8.doi:10.1074/jbc.M104323200.PMID11571281.
  82. ^Ponzetto C, Zhen Z, Audero E, Maina F, Bardelli A, Basile ML, Giordano S, Narsimhan R, Comoglio P (1996)."Specific uncoupling of GRB2 from the Met receptor. Differential effects on transformation and motility".J. Biol. Chem.271(24): 14119–23.doi:10.1074/jbc.271.24.14119.PMID8662889.
  83. ^Liang Q, Mohan RR, Chen L, Wilson SE (1998). "Signaling by HGF and KGF in corneal epithelial cells: Ras/MAP kinase and Jak-STAT pathways".Invest. Ophthalmol. Vis. Sci.39(8): 1329–38.PMID9660480.
  84. ^Comoglio PM (1993). "Structure, biosynthesis and biochemical properties of the HGF receptor in normal and malignant cells".EXS.65:131–65.PMID8380735.
  85. ^Naldini L, Weidner KM, Vigna E, Gaudino G, Bardelli A, Ponzetto C, Narsimhan RP, Hartmann G, Zarnegar R, Michalopoulos GK (1991)."Scatter factor and hepatocyte growth factor are indistinguishable ligands for the MET receptor".EMBO J.10(10): 2867–78.doi:10.1002/j.1460-2075.1991.tb07836.x.PMC452997.PMID1655405.
  86. ^Hiscox S, Jiang WG (1999). "Association of the HGF/SF receptor, c-met, with the cell-surface adhesion molecule, E-cadherin, and catenins in human tumor cells".Biochem. Biophys. Res. Commun.261(2): 406–11.doi:10.1006/bbrc.1999.1002.PMID10425198.
  87. ^Wang D, Li Z, Messing EM, Wu G (2002)."Activation of Ras/Erk pathway by a novel MET-interacting protein RanBPM".J. Biol. Chem.277(39): 36216–22.doi:10.1074/jbc.M205111200.PMID12147692.

Further reading

[edit]
[edit]