Hepatocyte growth factor receptor

Last updated

MET
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases MET , MET proto-oncogene, receptor tyrosine kinase, AUTS9, HGFR, RCCP2, c-Met, DFNB97, OSFD, c-met
External IDs OMIM: 164860 MGI: 96969 HomoloGene: 206 GeneCards: MET
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 Mb Chr 6: 17.46 – 17.57 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Hepatocyte growth factor receptor (HGF receptor) [5] [6] is a protein that in humans is encoded by the MET gene. The protein possesses tyrosine kinase activity. [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 of epithelial origin, while expression of HGF/SF is restricted to cells of mesenchymal origin. 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, only stem cells and progenitor cells express 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 cells are 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 its autocrine activation by co-expression of its hepatocyte growth factor ligand, have been implicated in oncogenesis. [8] [9]

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

Gene

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

Protein

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

Extracellular

Intracellular

A juxtamembrane segment that contains:

MET signaling pathway

MET activation by its ligand HGF induces 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 as GRB2, 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 high avidity, but low affinity. [21] Upon interaction with MET, GAB1 becomes phosphorylated on several tyrosine residues which, in turn, recruit a number of signalling effectors, including PI3K, 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

MET engagement activates multiple signal transduction pathways:

Role in development

MET mediates a complex program known as invasive growth. [26] Activation of MET triggers mitogenesis, and morphogenesis. [31] [32]

During embryonic development, transformation of the flat, two-layer germinal disc into a three-dimensional body depends on transition of some cells from an epithelial phenotype to spindle-shaped cells with motile behaviour, a mesenchymal phenotype. This process is referred to as epithelial-mesenchymal transition (EMT). [33] Later in embryonic development, MET is crucial for gastrulation, angiogenesis, myoblast migration, bone remodeling, and nerve sprouting among others. [34] MET is essential for embryogenesis, because MET−/− mice die in utero due to severe defects in placental development. [35] Along with Ectodysplasin 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 as liver regeneration and wound healing during adulthood. [26]

HGF/MET axis is also involved in myocardial development. Both HGF and MET receptor mRNAs are co-expressed in cardiomyocytes from 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 the endocardial cushion occurs. [38] However, MET is not essential for heart development, since α-MHCMet-KO mice show normal heart development. [39]

Expression

Tissue distribution

MET is normally expressed by epithelial cells. [26] However, MET is also found on endothelial cells, neurons, hepatocytes, hematopoietic cells, melanocytes and neonatal cardiomyocytes. [32] [40] HGF expression is restricted to cells of mesenchymal origin. [33]

Transcriptional control

MET transcription is activated by HGF and several growth factors. [41] MET promoter has four putative binding sites for Ets, a family of transcription factors that control several invasive growth genes. [41] ETS1 activates MET transcription in vitro. [42] MET transcription is activated by hypoxia-inducible factor 1 (HIF1), which is activated by low concentration of intracellular oxygen. [43] HIF1 can bind to one of the several hypoxia response elements (HREs) in the MET promoter. [33] Hypoxia also activates transcription factor AP-1, which is involved in MET transcription. [33]

Clinical significance

Role in cancer

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) by miR-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 the clear cell tumor subtype. [46]

The amplification of the cell surface receptor MET often drives resistance to anti-EGFR therapies in colorectal cancer. [47]

Role in autism

The SFARIgene database lists MET with an autism score 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 of schizophrenia. 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

In adult mice, MET is required to protect cardiomyocytes by preventing age-related oxidative stress, apoptosis, fibrosis and cardiac dysfunction. [39] Moreover, MET inhibitors, such as Crizotinib or 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 of caspases, metabolism alteration and blockage of ion 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 of cardiac stem cells. [59] [60]

Interaction with tumour suppressor genes

PTEN

PTEN (phosphatase and tensin homolog) is a tumor suppressor gene encoding a protein PTEN, which possesses lipid and protein phosphatase-dependent as well as phosphatase-independent activities. [61] PTEN protein phosphatase is able to interfere with MET signaling by dephosphorylating either PIP3 generated by PI3K, or the p52 isoform of SHC. SHC dephosphorylation inhibits recruitment of the GRB2 adapter to activated MET. [29]

VHL

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

Cancer therapies targeting HGF/MET

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 found here.

MET kinase inhibitors

Kinase inhibitors are low molecular weight molecules that prevent ATP binding 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.

HGF inhibitors

Since HGF is the only known ligand of MET, blocking the formation of a HGF:MET complex blocks MET biological 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.

Decoy MET

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 preventing metastasis in animal models. [72]

Immunotherapy targeting MET

Drugs used for immunotherapy can act either passively by enhancing the immunologic response to MET-expressing tumor cells, or actively by stimulating immune cells and altering differentiation/growth of tumor cells. [73]

Passive immunotherapy

Administering monoclonal antibodies (mAbs) is a form of passive immunotherapy. MAbs facilitate destruction of tumor cells by complement-dependent cytotoxicity (CDC) and cell-mediated cytotoxicity (ADCC). In CDC, mAbs bind to specific antigen, leading to activation of the complement cascade, which in turn leads to formation of pores in tumor cells. In ADCC, the Fab domain of a mAb binds to a tumor antigen, and Fc domain binds to Fc receptors present on effector cells (phagocytes and NK cells), thus forming a bridge between an effector and a target cells. This induces the effector cell activation, leading to phagocytosis of the tumor cell by neutrophils and macrophages. Furthermore, NK cells release cytotoxic molecules, which lyse tumor cells. [73]

  • DN30 is monoclonal anti-MET antibody that recognizes the extracellular portion of MET. DN30 induces both shedding of the MET ectodomain as well as cleavage of the intracellular domain, which is successively degraded by proteasome machinery. 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 prevents metastasis in animal models. [74]
  • OA-5D5 is one-armed monoclonal anti-MET antibody that was demonstrated to inhibit orthotopic pancreatic [75] and glioblastoma [76] tumor growth and to improve survival in tumor xenograft models. OA-5D5 is produced as a recombinant protein in Escherichia coli. It is composed of murine variable domains for the heavy and light chains with human IgG1 constant domains. The antibody blocks HGF binding to MET in a competitive fashion.

Active immunotherapy

Active immunotherapy to MET-expressing tumors can be achieved by administering cytokines, such as interferons (IFNs) and interleukins (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 the U.S. Food and Drug Administration (FDA) for the treatment of renal cell carcinoma and metastatic melanoma, which often have deregulated MET activity. [73]

Interactions

Met has been shown to interact with:

See also

Related Research Articles

<span class="mw-page-title-main">Tyrosine kinase</span> Class hi residues

A tyrosine kinase is an enzyme that can transfer a phosphate group from ATP to the tyrosine residues of specific proteins inside a cell. It functions as an "on" or "off" switch in many cellular functions.

<span class="mw-page-title-main">Protein kinase B</span> Set of three serine/threonine-specific protein kinases

Protein kinase B (PKB), also known as Akt, is the collective name of a set of three serine/threonine-specific protein kinases that play key roles in multiple cellular processes such as glucose metabolism, apoptosis, cell proliferation, transcription, and cell migration.

<span class="mw-page-title-main">Epidermal growth factor receptor</span> Transmembrane protein

The epidermal growth factor receptor is a transmembrane protein that is a receptor for members of the epidermal growth factor family of extracellular protein ligands.

<span class="mw-page-title-main">Tropomyosin receptor kinase A</span> Protein-coding gene in the species Homo sapiens

Tropomyosin receptor kinase A (TrkA), also known as high affinity nerve growth factor receptor, neurotrophic tyrosine kinase receptor type 1, or TRK1-transforming tyrosine kinase protein is a protein that in humans is encoded by the NTRK1 gene.

<span class="mw-page-title-main">Receptor tyrosine kinase</span> Class of enzymes

Receptor tyrosine kinases (RTKs) are the high-affinity cell surface receptors for many polypeptide growth factors, cytokines, and hormones. Of the 90 unique tyrosine kinase genes identified in the human genome, 58 encode receptor tyrosine kinase proteins. Receptor tyrosine kinases have been shown not only to be key regulators of normal cellular processes but also to have a critical role in the development and progression of many types of cancer. Mutations in receptor tyrosine kinases lead to activation of a series of signalling cascades which have numerous effects on protein expression. Receptor tyrosine kinases are part of the larger family of protein tyrosine kinases, encompassing the receptor tyrosine kinase proteins which contain a transmembrane domain, as well as the non-receptor tyrosine kinases which do not possess transmembrane domains.

<span class="mw-page-title-main">GRB2</span> Protein-coding gene in the species Homo sapiens

Growth factor receptor-bound protein 2, also known as Grb2, is an adaptor protein involved in signal transduction/cell communication. In humans, the GRB2 protein is encoded by the GRB2 gene.

<span class="mw-page-title-main">Hepatocyte growth factor</span> Mammalian protein found in Homo sapiens

Hepatocyte growth factor (HGF) or scatter factor (SF) is a paracrine cellular growth, motility and morphogenic factor. It is secreted by mesenchymal cells and targets and acts primarily upon epithelial cells and endothelial cells, but also acts on haemopoietic progenitor cells and T cells. It has been shown to have a major role in embryonic organ development, specifically in myogenesis, in adult organ regeneration, and in wound healing.

<span class="mw-page-title-main">KIT (gene)</span> Mammalian protein and protein-coding gene

Proto-oncogene c-KIT is the gene encoding the receptor tyrosine kinase protein known as tyrosine-protein kinase KIT, CD117 or mast/stem cell growth factor receptor (SCFR). Multiple transcript variants encoding different isoforms have been found for this gene. KIT was first described by the German biochemist Axel Ullrich in 1987 as the cellular homolog of the feline sarcoma viral oncogene v-kit.

The ErbB family of proteins contains four receptor tyrosine kinases, structurally related to the epidermal growth factor receptor (EGFR), its first discovered member. In humans, the family includes Her1, Her2 (ErbB2), Her3 (ErbB3), and Her4 (ErbB4). The gene symbol, ErbB, is derived from the name of a viral oncogene to which these receptors are homologous: erythroblastic leukemia viral oncogene. Insufficient ErbB signaling in humans is associated with the development of neurodegenerative diseases, such as multiple sclerosis and Alzheimer's disease, while excessive ErbB signaling is associated with the development of a wide variety of types of solid tumor.

<span class="mw-page-title-main">FYN</span> Mammalian protein found in Homo sapiens

Proto-oncogene tyrosine-protein kinase Fyn is an enzyme that in humans is encoded by the FYN gene.

<span class="mw-page-title-main">ERBB3</span> Protein found in humans

Receptor tyrosine-protein kinase erbB-3, also known as HER3, is a membrane bound protein that in humans is encoded by the ERBB3 gene.

<span class="mw-page-title-main">GRB2-associated-binding protein 1</span> Protein-coding gene in the species Homo sapiens

GRB2-associated-binding protein 1 is a protein that in humans is encoded by the GAB1 gene.

<span class="mw-page-title-main">SPRY2</span> Protein-coding gene in the species Homo sapiens

Sprouty homolog 2 (Drosophila), also known as SPRY2, is a protein which in humans is encoded by the SPRY2 gene.

<span class="mw-page-title-main">Proto-oncogene tyrosine-protein kinase Src</span> Mammalian protein found in Homo sapiens

Proto-oncogene tyrosine-protein kinase Src, also known as proto-oncogene c-Src, or simply c-Src, is a non-receptor tyrosine kinase protein that in humans is encoded by the SRC gene. It belongs to a family of Src family kinases and is similar to the v-Src gene of Rous sarcoma virus. It includes an SH2 domain, an SH3 domain and a tyrosine kinase domain. Two transcript variants encoding the same protein have been found for this gene.

<span class="mw-page-title-main">AXL receptor tyrosine kinase</span> Protein-coding gene in the species Homo sapiens

Tyrosine-protein kinase receptor UFO is an enzyme that in humans is encoded by the AXL gene. The gene was initially designated as UFO, in allusion to the unidentified function of this protein. However, in the years since its discovery, research into AXL's expression profile and mechanism has made it an increasingly attractive target, especially for cancer therapeutics. In recent years, AXL has emerged as a key facilitator of immune escape and drug-resistance by cancer cells, leading to aggressive and metastatic cancers.

<span class="mw-page-title-main">MST1R</span> Protein-coding gene in the species Homo sapiens

Macrophage-stimulating protein receptor is a protein that in humans is encoded by the MST1R gene. MST1R is also known as RON kinase, named after the French city in which it was discovered. It is related to the c-MET receptor tyrosine kinase.

<span class="mw-page-title-main">ROS1</span> Protein-coding gene in the species Homo sapiens

Proto-oncogene tyrosine-protein kinase ROS is an enzyme that in humans is encoded by the ROS1 gene.

Src kinase family is a family of non-receptor tyrosine kinases that includes nine members: Src, Yes, Fyn, and Fgr, forming the SrcA subfamily, Lck, Hck, Blk, and Lyn in the SrcB subfamily, and Frk in its own subfamily. Frk has homologs in invertebrates such as flies and worms, and Src homologs exist in organisms as diverse as unicellular choanoflagellates, but the SrcA and SrcB subfamilies are specific to vertebrates. Src family kinases contain six conserved domains: a N-terminal myristoylated segment, a SH2 domain, a SH3 domain, a linker region, a tyrosine kinase domain, and C-terminal tail.

c-Met inhibitors are a class of small molecules that inhibit the enzymatic activity of the c-Met tyrosine kinase, the receptor of hepatocyte growth factor/scatter factor (HGF/SF). These inhibitors may have therapeutic application in the treatment of various types of cancers.

George K. Michalopoulos is a Greek-American pathologist and academic. He served as Maud L. Menten Professor of Experimental Pathology and Chair of the Department of Pathology at the University of Pittsburgh and UPMC from 1991 to 2023.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000105976 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000009376 - Ensembl, 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. PMID   1846706.
  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. PMID   1611909.
  7. Cooper CS (January 1992). "The met oncogene: from detection by transfection to transmembrane receptor for hepatocyte growth factor". Oncogene. 7 (1): 3–7. PMID   1531516.
  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. PMID   7553606.
  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. PMID   8930021.
  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. PMID   4069211. S2CID   4359961.
  12. 1 2 Birchmeier 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. PMID   14685170. S2CID   19330786.
  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 . PMID   8294430.
  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 . PMID   11741535.
  15. 1 2 Ponzetto 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. PMID   7513258. S2CID   23383203.
  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 . PMID   8898205. S2CID   12943699.
  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. PMID   7549943.
  18. 1 2 Pelicci 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. PMID   7731718.
  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. PMID   8906793. S2CID   4357372.
  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. PMID   11114738. S2CID   22385297.
  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. PMID   11313945. S2CID   35447713.
  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. PMID   10734310. S2CID   22727382.
  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 . PMID   15239951.
  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 . PMID   7834738. S2CID   8995643.
  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 . PMID   1718989.
  26. 1 2 3 4 Gentile 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. PMID   18175071. S2CID   33076010.
  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. PMID   9440692. S2CID   30330705.
  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. PMID   11929826.
  29. 1 2 Abounader 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. PMID   15516982. S2CID   7623249.
  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. PMC   3760732 . PMID   18369158.
  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 . PMID   8514254.
  32. 1 2 "he fields of HGF/c-Met involvement". HealthValue. Archived from the original on 27 September 2007. Retrieved 13 June 2009.
  33. 1 2 3 4 Boccaccio 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. PMID   16862193. S2CID   396385.
  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. PMID   9789329.
  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. PMID   7854453. S2CID   4361262.
  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. PMC   4931016 . PMID   27336951.
  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. PMID   8635233.
  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 . PMID   9915579.
  39. 1 2 Arechederra 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 . PMID   23994610.
  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 . PMC   3036588 . PMID   21347410.
  41. 1 2 Shirasaki 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 . PMID   10618716.
  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. PMID   8934537.
  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 . PMID   12726861.
  44. "HGF/c-Met and cancer". HealthValue. Archived from the original on 27 September 2007. Retrieved 13 June 2009.
  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 . PMID   18456660.
  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. PMID   22525820.
  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. PMC   4078408 . PMID   23729478.
  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 . PMC   1838551 . PMID   17053076.
  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. PMID   19255034. S2CID   5395283.
  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. PMID   24909855. S2CID   5608613.
  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. PMC   4252539 . PMID   25471559.
  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. PMC   3942873 . PMID   23787772.
  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. PMC   3261279 . PMID   21509596.
  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. PMC   3086026 . PMID   21490227.
  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. PMC   2952412 . PMID   20853516.
  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. PMC   3454529 . PMID   22958829.
  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. PMID   23707608.
  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 . PMID   20231546.
  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. PMC   1519349 . PMID   2040247.
  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. PMID   21327916. S2CID   32535928.
  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 . PMID   9593664.
  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 . PMID   15930277.
  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. PMC   2423296 . PMID   18285456.
  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. PMID   12118367. S2CID   32305287.
  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. PMID   15064724. S2CID   12545344.
  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. PMID   14617781.
  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. PMID   14612533.
  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 . PMC   1413705 . PMID   16461907.
  69. 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. PMID   12824898. S2CID   24806218.
  70. 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 . PMC   34688 . PMID   11416216.
  71. 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. PMID   16452232.
  72. 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 . PMID   15261142.
  73. 1 2 3 Reang P, Gupta M, Kohli K (2006). "Biological response modifiers in cancer". MedGenMed. 8 (4): 33. PMC   1868326 . PMID   17415315.
  74. 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 . PMC   1458799 . PMID   16547140.
  75. 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 . PMID   18519697.
  76. 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 . PMID   17062691.
  77. 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. PMID   11254878.
  78. 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. PMID   11894096. S2CID   4389099.
  79. 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. PMC   2265755 . PMID   18273061.
  80. 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 . PMID   11571281.
  81. 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 . PMID   8662889.
  82. 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. PMID   9660480.
  83. Comoglio PM (1993). "Structure, biosynthesis and biochemical properties of the HGF receptor in normal and malignant cells". EXS. 65: 131–65. PMID   8380735.
  84. 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. PMC   452997 . PMID   1655405.
  85. 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. PMID   10425198.
  86. 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 . PMID   12147692.

Further reading