ijms-logo

Journal Browser

Journal Browser

Hepatocyte Growth Factor (HGF), II

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Pathology, Diagnostics, and Therapeutics".

Deadline for manuscript submissions: closed (30 September 2020) | Viewed by 42284

Special Issue Editors


E-Mail Website
Guest Editor
Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan
Interests: HGF expression; HGF function; antitumor immune response; immune tolerance; neuronal differentiation
Special Issues, Collections and Topics in MDPI journals

E-Mail
Guest Editor
Department of HGF liver regeneration and tissue repair, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8520, Japan and Kagoshima City Hospital, Kagoshima 890-8760, Japan
Interests: HGF pathophysiology; HGF therapeutic application; liver regeneration

Special Issue Information

Dear Colleagues,

This Special Issue is the continuation of our previous Special Issue "Hepatocyte Growth Factor (HGF)".

Hepatocyte growth factor (HGF), also known as hepatopoietin-A (HPTA), scatter factor (SF), and fibroblast-derived tumor cytotoxic factor (F-TCF), is a pleiotropic growth factor that is secreted mainly from mesenchymal cells and acts on epithelial cells, endothelial cells, and other types of cells. The receptor of HGF is the product of c-met proto-oncogene with tyrosine kinase activity that mediates the transduction of multiple biological signals of HGF. Since HGF was purified and HGF cDNA was molecularly cloned in the late 1980s, extensive studies have been carried out in the fields of not only tissue regeneration but also cell biology, tumor biology, neurology, immunology, and others. 

This Special Issue of the International Journal of Molecular Sciences will focus on recent advances in “Hepatocyte Growth Factor (HGF)” research, including new insights into liver regeneration, activation, biological and pharmacological activities, signaling pathways, structure-function relationships, regulation of expression, therapeutic and diagnostic applications, and other aspects of HGF. Original research papers and review articles relevant to those topics are welcome.

You may wish to check the previous volume.

Prof. Dr. Eiichi Gohda
Prof. Dr. Hirohito Tsubouchi
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Related Special Issue

Published Papers (24 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

14 pages, 3107 KiB  
Article
Dimer Interface in Natural Variant NK1 Is Dispensable for HGF-Dependent Met Receptor Activation
by Yumiko Tahira, Katsuya Sakai, Hiroki Sato, Ryu Imamura and Kunio Matsumoto
Int. J. Mol. Sci. 2021, 22(17), 9240; https://doi.org/10.3390/ijms22179240 - 26 Aug 2021
Cited by 1 | Viewed by 2155
Abstract
NK1, a splicing variant of hepatocyte growth factor (HGF), binds to and activates Met receptor by forming an NK1 dimer and 2:2 complex with Met. Although the structural mechanism underlying Met activation by HGF remains incompletely resolved, it has been proposed that the [...] Read more.
NK1, a splicing variant of hepatocyte growth factor (HGF), binds to and activates Met receptor by forming an NK1 dimer and 2:2 complex with Met. Although the structural mechanism underlying Met activation by HGF remains incompletely resolved, it has been proposed that the NK1 dimer structure participates in this activation. We investigated the NK1 dimer interface’s role in Met activation by HGF. Because N127, V140, and K144 are closely involved in the head-to-tail NK1 dimer formation, mutant NK1 proteins with replacement of these residues by alanine were prepared. In Met tyrosine phosphorylation assays, N127-NK1, V140-NK1, and K144-NK1 showed 8.3%, 23.8%, and 52.2% activity, respectively, compared with wild-type NK1. Although wild-type NK1 promoted cell migration and scattering, N127-NK1, V140-NK1, and K144-NK1 hardly or marginally promoted them, indicating loss of activity of these mutant NK1 proteins to activate Met. In contrast, mutant HGFs (N127-HGF, V140-HGF, and K144-HGF) with the same amino acid replacements as in NK1 induced Met tyrosine phosphorylation and biological responses at levels comparable to those of wild-type HGF. These results indicate that the structural basis responsible for NK1-dependent Met dimer formation and activation differs from, or is at least distinguishable from, the structural basis responsible for HGF-dependent Met activation. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

11 pages, 1096 KiB  
Article
Serum APOA4 Pharmacodynamically Represents Administered Recombinant Human Hepatocyte Growth Factor (E3112)
by Sotaro Motoi, Mai Uesugi, Takashi Obara, Katsuhiro Moriya, Yoshihisa Arita, Hideaki Ogasawara, Motohiro Soejima, Toshio Imai and Tetsu Kawano
Int. J. Mol. Sci. 2021, 22(9), 4578; https://doi.org/10.3390/ijms22094578 - 27 Apr 2021
Cited by 2 | Viewed by 2042
Abstract
Background: Hepatocyte growth factor (HGF) is an endogenously induced bioactive molecule that has strong anti-apoptotic and tissue repair activities. In this research, we identified APOA4 as a novel pharmacodynamic (PD) marker of the recombinant human HGF (rh-HGF), E3112. Methods: rh-HGF was administered to [...] Read more.
Background: Hepatocyte growth factor (HGF) is an endogenously induced bioactive molecule that has strong anti-apoptotic and tissue repair activities. In this research, we identified APOA4 as a novel pharmacodynamic (PD) marker of the recombinant human HGF (rh-HGF), E3112. Methods: rh-HGF was administered to mice, and their livers were investigated for the PD marker. Candidates were identified from soluble proteins and validated by using human hepatocytes in vitro and an animal disease model in vivo, in which its c-Met dependency was also ensured. Results: Among the genes induced or highly enhanced after rh-HGF exposure in vivo, a soluble apolipoprotein, Apoa4, was found to be induced by rh-HGF in the murine liver. By using primary cultured human hepatocytes, the significant induction of human APOA4 was observed at the mRNA and protein levels, and it was inhibited in the presence of a c-Met inhibitor. Although mice constitutively expressed Apoa4 mRNA in the small intestine and the liver, the liver was the primary organ affected by administered rh-HGF to strongly induce APOA4 in a dose- and c-Met-dependent manner. Serum APOA4 levels were increased after rh-HGF administration, not only in normal mice but also in anti-Fas-induced murine acute liver failure (ALF), which confirmed the pharmacodynamic nature of APOA4. Conclusions: APOA4 was identified as a soluble PD marker of rh-HGF with c-Met dependency. It should be worthwhile to clinically validate its utility through clinical trials with healthy subjects and ALF patients. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

21 pages, 5145 KiB  
Article
The PI3K/AKT Pathway Is Activated by HGF in NT2D1 Non-Seminoma Cells and Has a Role in the Modulation of Their Malignant Behavior
by Luisa Gesualdi, Erica Leonetti, Alessandra Cucina, Bianca Maria Scicchitano, Silvia Sorrentino, Maria Grazia Tarsitano, Andrea Isidori, Mariano Bizzarri, Antonio Filippini, Anna Riccioli, Marcella Cammarota, Vincenzo Gigantino, Giulia Ricci and Angela Catizone
Int. J. Mol. Sci. 2020, 21(22), 8669; https://doi.org/10.3390/ijms21228669 - 17 Nov 2020
Cited by 6 | Viewed by 1891
Abstract
Overactivation of the c-MET/HGF system is a feature of many cancers. We previously reported that type II testicular germ cell tumor (TGCT) cells express the c-MET receptor, forming non-seminomatous lesions that are more positive compared with seminomatous ones. Notably, we also demonstrated that [...] Read more.
Overactivation of the c-MET/HGF system is a feature of many cancers. We previously reported that type II testicular germ cell tumor (TGCT) cells express the c-MET receptor, forming non-seminomatous lesions that are more positive compared with seminomatous ones. Notably, we also demonstrated that NT2D1 non-seminomatous cells (derived from an embryonal carcinoma lesion) increase their proliferation, migration, and invasion in response to HGF. Herein, we report that HGF immunoreactivity is more evident in the microenvironment of embryonal carcinoma biopsies with respect to seminomatous ones, indicating a tumor-dependent modulation of the testicular niche. PI3K/AKT is one of the signaling pathways triggered by HGF through the c-MET activation cascade. Herein, we demonstrated that phospho-AKT increases in NT2D1 cells after HGF stimulation. Moreover, we found that this pathway is involved in HGF-dependent NT2D1 cell proliferation, migration, and invasion, since the co-administration of the PI3K inhibitor LY294002 together with HGF abrogates these responses. Notably, the inhibition of endogenous PI3K affects collective cell migration but does not influence proliferation or chemotactic activity. Surprisingly, LY294002 administered without the co-administration of HGF increases cell invasion at levels comparable to the HGF-administered samples. This paradoxical result highlights the role of the testicular microenvironment in the modulation of cellular responses and stimulates the study of the testicular secretome in cancer lesions. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

17 pages, 4360 KiB  
Article
Transplantation of Adipose Stromal Cell Sheet Producing Hepatocyte Growth Factor Induces Pleiotropic Effect in Ischemic Skeletal Muscle
by Maria A. Boldyreva, Evgeny K. Shevchenko, Yuliya D. Molokotina, Pavel I. Makarevich, Irina B. Beloglazova, Ekaterina S. Zubkova, Konstantin V. Dergilev, Zoya I. Tsokolaeva, Dmitry Penkov, Mu-Nung Hsu, Yu-Chen Hu and Yelena V. Parfyonova
Int. J. Mol. Sci. 2019, 20(12), 3088; https://doi.org/10.3390/ijms20123088 - 24 Jun 2019
Cited by 19 | Viewed by 5111
Abstract
Cell therapy remains a promising approach for the treatment of cardiovascular diseases. In this regard, the contemporary trend is the development of methods to overcome low cell viability and enhance their regenerative potential. In the present study, we evaluated the therapeutic potential of [...] Read more.
Cell therapy remains a promising approach for the treatment of cardiovascular diseases. In this regard, the contemporary trend is the development of methods to overcome low cell viability and enhance their regenerative potential. In the present study, we evaluated the therapeutic potential of gene-modified adipose-derived stromal cells (ADSC) that overexpress hepatocyte growth factor (HGF) in a mice hind limb ischemia model. Angiogenic and neuroprotective effects were assessed following ADSC transplantation in suspension or in the form of cell sheet. We found superior blood flow restoration, tissue vascularization and innervation, and fibrosis reduction after transplantation of HGF-producing ADSC sheet compared to other groups. We suggest that the observed effects are determined by pleiotropic effects of HGF, along with the multifactorial paracrine action of ADSC which remain viable and functionally active within the engineered cell construct. Thus, we demonstrated the high therapeutic potential of the utilized approach for skeletal muscle recovery after ischemic damage associated with complex tissue degenerative effects. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

14 pages, 10825 KiB  
Article
Distinct Localization of Mature HGF from its Precursor Form in Developing and Repairing the Stomach
by Nawaphat Jangphattananont, Hiroki Sato, Ryu Imamura, Katsuya Sakai, Yumi Terakado, Kazuhiro Murakami, Nick Barker, Hiroko Oshima, Masanobu Oshima, Junichi Takagi, Yukinari Kato, Seiji Yano and Kunio Matsumoto
Int. J. Mol. Sci. 2019, 20(12), 2955; https://doi.org/10.3390/ijms20122955 - 17 Jun 2019
Cited by 9 | Viewed by 5987
Abstract
Hepatocyte growth factor (HGF) is secreted as an inactive single-chain HGF (scHGF); however, only proteolytically processed two-chain HGF (tcHGF) can activate the MET receptor. We investigated the localization of tcHGF and activated/phosphorylated MET (pMET) using a tcHGF-specific antibody. In day 16.5 mouse embryos, [...] Read more.
Hepatocyte growth factor (HGF) is secreted as an inactive single-chain HGF (scHGF); however, only proteolytically processed two-chain HGF (tcHGF) can activate the MET receptor. We investigated the localization of tcHGF and activated/phosphorylated MET (pMET) using a tcHGF-specific antibody. In day 16.5 mouse embryos, total HGF (scHGF + tcHGF) was mainly localized in smooth muscle cells close to, but separate from, MET-positive epithelial cells in endodermal organs, including the stomach. In the adult stomach, total HGF was localized in smooth muscle cells, and tcHGF was mainly localized in the glandular base region. Immunostaining for pMET and Lgr5-driven green fluorescent protein (GFP) indicated that pMET localization overlapped with Lgr5+ gastric stem cells. HGF promoted organoid formation similar to EGF, indicating the potential for HGF to promote the survival and growth of gastric stem cells. pMET and tcHGF localizations changed during regeneration following gastric injury. These results indicate that MET is constantly activated in gastric stem cells and that the localization of pMET differs from the primary localization of precursor HGF but has a close relationship to tcHGF. Our results suggest the importance of the microenvironmental generation of tcHGF in the regulation of development, regeneration, and stem cell behavior. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

9 pages, 4530 KiB  
Article
Anti-Apoptotic Effects of Recombinant Human Hepatocyte Growth Factor on Hepatocytes Were Associated with Intrahepatic Hemorrhage Suppression Indicated by the Preservation of Prothrombin Time
by Sotaro Motoi, Hiroko Toyoda, Takashi Obara, Etsuko Ohta, Yoshihisa Arita, Kana Negishi, Katsuhiro Moriya, Yoshikazu Kuboi, Motohiro Soejima, Toshio Imai, Akio Ido, Hirohito Tsubouchi and Tetsu Kawano
Int. J. Mol. Sci. 2019, 20(8), 1821; https://doi.org/10.3390/ijms20081821 - 12 Apr 2019
Cited by 13 | Viewed by 2783
Abstract
Hepatocyte growth factor (HGF) is an endogenously expressed bioactive substance that has a strong anti-apoptotic effect. In this study, we biochemically and histologically characterized the effects of rh-HGF on in vitro human hepatocyte injury and mouse acute liver failure (ALF) models, both of [...] Read more.
Hepatocyte growth factor (HGF) is an endogenously expressed bioactive substance that has a strong anti-apoptotic effect. In this study, we biochemically and histologically characterized the effects of rh-HGF on in vitro human hepatocyte injury and mouse acute liver failure (ALF) models, both of which were induced by antibody-mediated Fas signaling. rh-HGF inhibited intracellular caspase-3/7 activation and cytokeratin 18 (CK-18) fragment release in both models. Histologically, rh-HGF dramatically suppressed parenchymal damage and intrahepatic hemorrhage. Among the laboratory parameters, prothrombin time (PT) was strongly preserved by rh-HGF, and PT was well correlated with the degree of intrahepatic hemorrhage. These results showed that the anti-apoptotic effect of rh-HGF on hepatocytes coincided strikingly with the suppression of intrahepatic hemorrhage. PT was considered to be the best parameter that correlated with the intrahepatic hemorrhages associated with hepatocellular damage. The action of rh-HGF might derive not only from its anti-apoptosis effects on liver parenchymal cells but also from its stabilization of structural and vasculature integrity. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

11 pages, 1198 KiB  
Communication
Efficient Selection of Antibodies Reactive to Homologous Epitopes on Human and Mouse Hepatocyte Growth Factors by Next-Generation Sequencing-Based Analysis of the B Cell Repertoire
by Soohyun Kim, Hyunho Lee, Jinsung Noh, Yonghee Lee, Haejun Han, Duck Kyun Yoo, Hyori Kim, Sunghoon Kwon and Junho Chung
Int. J. Mol. Sci. 2019, 20(2), 417; https://doi.org/10.3390/ijms20020417 - 18 Jan 2019
Cited by 3 | Viewed by 4434
Abstract
YYB-101 is a humanized rabbit anti-human hepatocyte growth factor (HGF)-neutralizing antibody currently in clinical trial. To test the effect of HGF neutralization with antibody on anti-cancer T cell immunity, we generated surrogate antibodies that are reactive to the mouse homologue of the epitope [...] Read more.
YYB-101 is a humanized rabbit anti-human hepatocyte growth factor (HGF)-neutralizing antibody currently in clinical trial. To test the effect of HGF neutralization with antibody on anti-cancer T cell immunity, we generated surrogate antibodies that are reactive to the mouse homologue of the epitope targeted by YYB-101. First, we immunized a chicken with human HGF and monitored changes in the B cell repertoire by next-generation sequencing (NGS). We then extracted the VH gene repertoire from the NGS data, clustered it into components by sequence homology, and classified the components by the change in the number of unique VH sequences and the frequencies of the VH sequences within each component following immunization. Those changes should accompany the preferential proliferation and somatic hypermutation or gene conversion of B cells encoding HGF-reactive antibodies. One component showed significant increases in the number and frequencies of unique VH sequences and harbored genes encoding antibodies that were reactive to human HGF and competitive with YYB-101 for HGF binding. Some of the antibodies also reacted to mouse HGF. The selected VH sequences shared 98.3% identity and 98.9% amino acid similarity. It is therefore likely that the antibodies encoded by them all react to the epitope targeted by YYB-101. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Figure 1

21 pages, 11802 KiB  
Article
c-Src Recruitment is Involved in c-MET-Mediated Malignant Behaviour of NT2D1 Non-Seminoma Cells
by Erica Leonetti, Luisa Gesualdi, Katia Corano Scheri, Simona Dinicola, Luigi Fattore, Maria Grazia Masiello, Alessandra Cucina, Rita Mancini, Mariano Bizzarri, Giulia Ricci and Angela Catizone
Int. J. Mol. Sci. 2019, 20(2), 320; https://doi.org/10.3390/ijms20020320 - 14 Jan 2019
Cited by 9 | Viewed by 3297
Abstract
c-MET pathway over-activation is the signature of malignancy acquisition or chemotherapy resistance of many cancers. We recently demonstrated that type II Testicular Germ Cell Tumours (TGCTs) express c-MET receptor. In particular, we elucidated that the non-seminoma lesions express c-MET protein at higher level, [...] Read more.
c-MET pathway over-activation is the signature of malignancy acquisition or chemotherapy resistance of many cancers. We recently demonstrated that type II Testicular Germ Cell Tumours (TGCTs) express c-MET receptor. In particular, we elucidated that the non-seminoma lesions express c-MET protein at higher level, compared with the seminoma ones. In line with this observation, NTERA-2 clone D1 (NT2D1) non-seminoma cells increase their proliferation, migration and invasion in response to Hepatocyte Growth Factor (HGF). One of the well-known adaptor-proteins belonging to c-MET signaling cascade is c-Src. Activation of c-Src is related to the increase of aggressiveness of many cancers. For this reason, we focused on the role of c-Src in c-MET-triggered and HGF-dependent NT2D1 cell activities. In the present paper, we have elucidated that this adaptor-protein is involved in HGF-dependent NT2D1 cell proliferation, migration and invasion, since Src inhibitor-1 administration abrogates these responses. Despite these biological evidences western blot analyses have not revealed the increase of c-Src activation because of HGF administration. However, notably, immunofluorescence analyses revealed that cytoplasmic and membrane-associated localization of c-Src shifted to the nuclear compartment after HGF stimulation. These results shed new light in the modality of HGF-dependent c-Src recruitment, and put the basis for novel investigations on the relationship between c-Src, and TGCT aggressiveness. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Graphical abstract

13 pages, 8289 KiB  
Article
Matriptase-Induced Phosphorylation of MET is Significantly Associated with Poor Prognosis in Invasive Bladder Cancer; an Immunohistochemical Analysis
by Koji Yamasaki, Shoichiro Mukai, Takahiro Nagai, Kozue Nakahara, Masato Fujii, Naoki Terada, Akinobu Ohno, Yuichiro Sato, Yoshinobu Toda, Hiroaki Kataoka and Toshiyuki Kamoto
Int. J. Mol. Sci. 2018, 19(12), 3708; https://doi.org/10.3390/ijms19123708 - 22 Nov 2018
Cited by 13 | Viewed by 3262
Abstract
Hepatocyte growth factor (HGF) plays an important role in cancer progression via phosphorylation of MET (c-met proto-oncogene product, receptor of HGF). HGF-zymogen (pro-HGF) must be processed for activation by HGF activators including matriptase, which is a type II transmembrane serine protease and [...] Read more.
Hepatocyte growth factor (HGF) plays an important role in cancer progression via phosphorylation of MET (c-met proto-oncogene product, receptor of HGF). HGF-zymogen (pro-HGF) must be processed for activation by HGF activators including matriptase, which is a type II transmembrane serine protease and the most efficient activator. The enzymatic activity is tightly regulated by HGF activator inhibitors (HAIs). Dysregulated pro-HGF activation (with upregulated MET phosphorylation) is reported to promote cancer progression in various cancers. We retrospectively analyzed the expression of matriptase, phosphorylated-MET (phospho-MET) and HAI-1 in tumor specimens obtained from patients with invasive bladder cancer by immunohistochemistry. High expression of phospho-MET and increased expression of matriptase were significantly associated with poor prognosis, and high matriptase/low HAI-1 expression showed poorer prognosis. Furthermore, high expression of matriptase tended to correlate with phosphorylation of MET. Increased expression of matriptase may induce the ligand-dependent activation of MET, which leads to poor prognosis in patients with invasive bladder cancer. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Graphical abstract

17 pages, 4507 KiB  
Article
MET Activation by a Macrocyclic Peptide Agonist that Couples to Biological Responses Differently from HGF in a Context-Dependent Manner
by Wenyu Miao, Katsuya Sakai, Ryu Imamura, Kenichiro Ito, Hiroaki Suga, Tetsushi Sakuma, Takashi Yamamoto and Kunio Matsumoto
Int. J. Mol. Sci. 2018, 19(10), 3141; https://doi.org/10.3390/ijms19103141 - 12 Oct 2018
Cited by 6 | Viewed by 4196
Abstract
Non-native ligands for growth factor receptors with distinct chemical properties and different biological activities have the potential to become therapeutic applications. We previously generated MET/hepatocyte growth factor (HGF) receptor agonists using bivalent macrocyclic peptides. The highest MET-activating agonists exhibited biological activity that was [...] Read more.
Non-native ligands for growth factor receptors with distinct chemical properties and different biological activities have the potential to become therapeutic applications. We previously generated MET/hepatocyte growth factor (HGF) receptor agonists using bivalent macrocyclic peptides. The highest MET-activating agonists exhibited biological activity that was indistinguishable from the effects of HGF. In this study, we investigated MET activation, signal characteristics, and biological responses induced by a macrocyclic peptide partial agonist known as aML5-PEG11. aML5-PEG11 induced weak tyrosine phosphorylation of MET while enhancing cell migration with potency comparable to HGF. aML5-PEG11 induced marked AKT (protein kinase B) and ERK (extracellular signal-regulated kinase) activation at a comparable potency and time-dependency to HGF, which suggests that enhancement of cell motility is attributable to activation of these molecules. In a 3-D culture of bile duct cancer cells in collagen gel, HGF induced robust activation of MET, ERK, and AKT, which was associated with enhanced expression of genes involved in bile duct development and subsequent branching of tubulogenesis. In contrast, aML5-PEG11 induced marginal activation of MET, ERK, and AKT (levels near the detection limits), which was associated with failure to enhance the expression of genes involved in bile duct development and a lack of tubulogenic response. Thus, MET activation by aML5-PEG11 couples to biological responses differently from HGF in an extracellular context-dependent manner. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Figure 1

Review

Jump to: Research

18 pages, 1614 KiB  
Review
Targeting HGF/c-MET Axis in Pancreatic Cancer
by Srinivasa P. Pothula, Zhihong Xu, David Goldstein, Romano C. Pirola, Jeremy S. Wilson and Minoti V. Apte
Int. J. Mol. Sci. 2020, 21(23), 9170; https://doi.org/10.3390/ijms21239170 - 01 Dec 2020
Cited by 35 | Viewed by 5169
Abstract
Pancreatic cancer (pancreatic ductal adenocarcinoma (PDAC/PC)) has been an aggressive disease that is associated with early metastases. It is characterized by dense and collagenous desmoplasia/stroma, predominantly produced by pancreatic stellate cells (PSCs). PSCs interact with cancer cells as well as other stromal cells, [...] Read more.
Pancreatic cancer (pancreatic ductal adenocarcinoma (PDAC/PC)) has been an aggressive disease that is associated with early metastases. It is characterized by dense and collagenous desmoplasia/stroma, predominantly produced by pancreatic stellate cells (PSCs). PSCs interact with cancer cells as well as other stromal cells, facilitating disease progression. A candidate growth factor pathway that may mediate this interaction is the hepatocyte growth factor (HGF)/c-MET pathway. HGF is produced by PSCs and its receptor c-MET is expressed on pancreatic cancer cells and endothelial cells. The current review discusses the role of the MET/HGF axis in tumour progression and dissemination of pancreatic cancer. Therapeutic approaches that were developed targeting either the ligand (HGF) or the receptor (c-MET) have not been shown to translate well into clinical settings. We discuss a two-pronged approach of targeting both the components of this pathway to interrupt the stromal–tumour interactions, which may represent a potential therapeutic strategy to improve outcomes in PC. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

11 pages, 690 KiB  
Review
The Potential Role of Hepatocyte Growth Factor in Degenerative Disorders of the Synovial Joint and Spine
by Hitoshi Tonomura, Masateru Nagae, Ryota Takatori, Hidenobu Ishibashi, Tomonori Itsuji and Kenji Takahashi
Int. J. Mol. Sci. 2020, 21(22), 8717; https://doi.org/10.3390/ijms21228717 - 18 Nov 2020
Cited by 16 | Viewed by 3140
Abstract
This paper aims to provide a comprehensive review of the changing role of hepatocyte growth factor (HGF) signaling in the healthy and diseased synovial joint and spine. HGF is a multifunctional growth factor that, like its specific receptor c-Met, is widely expressed in [...] Read more.
This paper aims to provide a comprehensive review of the changing role of hepatocyte growth factor (HGF) signaling in the healthy and diseased synovial joint and spine. HGF is a multifunctional growth factor that, like its specific receptor c-Met, is widely expressed in several bone and joint tissues. HGF has profound effects on cell survival and proliferation, matrix metabolism, inflammatory response, and neurotrophic action. HGF plays an important role in normal bone and cartilage turnover. Changes in HGF/c-Met have also been linked to pathophysiological changes in degenerative joint diseases, such as osteoarthritis (OA) and intervertebral disc degeneration (IDD). A therapeutic role of HGF has been proposed in the regeneration of osteoarticular tissues. HGF also influences bone remodeling and peripheral nerve activity. Studies aimed at elucidating the changing role of HGF/c-Met signaling in OA and IDD at different pathophysiological stages, and their specific molecular mechanisms are needed. Such studies will contribute to safe and effective HGF/c-Met signaling-based treatments for OA and IDD. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

15 pages, 5813 KiB  
Review
Cyclic Peptide-Based Biologics Regulating HGF-MET
by Hiroki Sato, Ryu Imamura, Hiroaki Suga, Kunio Matsumoto and Katsuya Sakai
Int. J. Mol. Sci. 2020, 21(21), 7977; https://doi.org/10.3390/ijms21217977 - 27 Oct 2020
Cited by 5 | Viewed by 3830
Abstract
Using a random non-standard peptide integrated discovery system, we obtained cyclic peptides that bind to hepatocyte growth factor (HGF) or mesenchymal-epithelial transition factor. (MET) HGF-inhibitory peptide-8 (HiP-8) selectively bound to two-chain active HGF, but not to single-chain precursor HGF. HGF showed a dynamic [...] Read more.
Using a random non-standard peptide integrated discovery system, we obtained cyclic peptides that bind to hepatocyte growth factor (HGF) or mesenchymal-epithelial transition factor. (MET) HGF-inhibitory peptide-8 (HiP-8) selectively bound to two-chain active HGF, but not to single-chain precursor HGF. HGF showed a dynamic change in its molecular shape in atomic force microscopy, but HiP-8 inhibited dynamic change in the molecular shape into a static status. The inhibition of the molecular dynamics of HGF by HiP-8 was associated with the loss of the ability to bind MET. HiP-8 could selectively detect active HGF in cancer tissues, and active HGF probed by HiP-8 showed co-localization with activated MET. Using HiP-8, cancer tissues with active HGF could be detected by positron emission tomography. HiP-8 seems to be applicable for the diagnosis and treatment of cancers. In contrast, based on the receptor dimerization as an essential process for activation, the cross-linking of the cyclic peptides that bind to the extracellular region of MET successfully generated an artificial ligand to MET. The synthetic MET agonists activated MET and exhibited biological activities which were indistinguishable from the effects of HGF. MET agonists composed of cyclic peptides can be manufactured by chemical synthesis but not recombinant protein expression, and thus are expected to be new biologics that are applicable to therapeutics and regenerative medicine. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

18 pages, 1651 KiB  
Review
HGF/MET Signaling in Malignant Brain Tumors
by Elizabeth Qian Xu Mulcahy, Rossymar Rivera Colόn and Roger Abounader
Int. J. Mol. Sci. 2020, 21(20), 7546; https://doi.org/10.3390/ijms21207546 - 13 Oct 2020
Cited by 21 | Viewed by 4041
Abstract
Hepatocyte growth factor (HGF) ligand and its receptor tyrosine kinase (RTK) mesenchymal-epithelial transition factor (MET) are important regulators of cellular processes such as proliferation, motility, angiogenesis, and tissue regeneration. In healthy adult somatic cells, this ligand and receptor pair is expressed at low [...] Read more.
Hepatocyte growth factor (HGF) ligand and its receptor tyrosine kinase (RTK) mesenchymal-epithelial transition factor (MET) are important regulators of cellular processes such as proliferation, motility, angiogenesis, and tissue regeneration. In healthy adult somatic cells, this ligand and receptor pair is expressed at low levels and has little activity except when tissue injuries arise. In cancer cells, HGF/MET are often overexpressed, and this overexpression is found to correlate with tumorigenesis, metastasis, and poorer overall prognosis. This review focuses on the signaling of these molecules in the context of malignant brain tumors. RTK signaling pathways are among the most common and universally dysregulated pathways in gliomas. We focus on the role of HGF/MET in the following primary malignant brain tumors: astrocytomas, glioblastomas, oligodendrogliomas, ependymomas, and embryonal central nervous system tumors (including medulloblastomas and others). Brain metastasis, as well as current advances in targeted therapies, are also discussed. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Graphical abstract

9 pages, 737 KiB  
Review
Multipotent Neurotrophic Effects of Hepatocyte Growth Factor in Spinal Cord Injury
by Kentaro Yamane, Haruo Misawa, Tomoyuki Takigawa, Yoshihiro Ito, Toshifumi Ozaki and Akihiro Matsukawa
Int. J. Mol. Sci. 2019, 20(23), 6078; https://doi.org/10.3390/ijms20236078 - 02 Dec 2019
Cited by 10 | Viewed by 4446
Abstract
Spinal cord injury (SCI) results in neural tissue loss and so far untreatable functional impairment. In addition, at the initial injury site, inflammation induces secondary damage, and glial scar formation occurs to limit inflammation-mediated tissue damage. Consequently, it obstructs neural regeneration. Many studies [...] Read more.
Spinal cord injury (SCI) results in neural tissue loss and so far untreatable functional impairment. In addition, at the initial injury site, inflammation induces secondary damage, and glial scar formation occurs to limit inflammation-mediated tissue damage. Consequently, it obstructs neural regeneration. Many studies have been conducted in the field of SCI; however, no satisfactory treatment has been established to date. Hepatocyte growth factor (HGF) is one of the neurotrophic growth factors and has been listed as a candidate medicine for SCI treatment. The highlighted effects of HGF on neural regeneration are associated with its anti-inflammatory and anti-fibrotic activities. Moreover, HGF exerts positive effects on transplanted stem cell differentiation into neurons. This paper reviews the mechanisms underlying the therapeutic effects of HGF in SCI recovery, and introduces recent advances in the clinical applications of HGF therapy. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF), II)
Show Figures

Figure 1

15 pages, 954 KiB  
Review
Application of Hepatocyte Growth Factor for Acute Spinal Cord Injury: The Road from Basic Studies to Human Treatment
by Kazuya Kitamura, Narihito Nagoshi, Osahiko Tsuji, Morio Matsumoto, Hideyuki Okano and Masaya Nakamura
Int. J. Mol. Sci. 2019, 20(5), 1054; https://doi.org/10.3390/ijms20051054 - 28 Feb 2019
Cited by 30 | Viewed by 5476
Abstract
Hepatocyte growth factor (HGF) was first identified as a potent mitogen for mature hepatocytes, and has also gained attention as a strong neurotrophic factor in the central nervous system. We found that during the acute phase of spinal cord injury (SCI) in rats, [...] Read more.
Hepatocyte growth factor (HGF) was first identified as a potent mitogen for mature hepatocytes, and has also gained attention as a strong neurotrophic factor in the central nervous system. We found that during the acute phase of spinal cord injury (SCI) in rats, c-Met, the specific receptor for HGF, increases sharply, while the endogenous HGF up-regulation is relatively weak. Introducing exogenous HGF into the spinal cord by injecting an HGF-expressing viral vector significantly increased the neuron and oligodendrocyte survival, angiogenesis, and axonal regeneration, to reduce the area of damage and to promote functional recovery in rats after SCI. Other recent studies in rodents have shown that exogenously administered HGF during the acute phase of SCI reduces astrocyte activation to decrease glial scar formation, and exerts anti-inflammatory effects to reduce leukocyte infiltration. We also reported that the intrathecal infusion of recombinant human HGF (intrathecal rhHGF) improves neurological hand function after cervical contusive SCI in the common marmoset, a non-human primate. Based on these collective results, we conducted a phase I/II clinical trial of intrathecal rhHGF for patients with acute cervical SCI who showed a modified Frankel grade of A/B1/B2 72 h after injury onset, from June 2014 to May 2018. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Graphical abstract

9 pages, 225 KiB  
Review
Targeting the Hepatocyte Growth Factor and c-Met Signaling Axis in Bone Metastases
by Young Mi Whang, Seung Pil Jung, Meyoung-Kon Kim, In Ho Chang and Serk In Park
Int. J. Mol. Sci. 2019, 20(2), 384; https://doi.org/10.3390/ijms20020384 - 17 Jan 2019
Cited by 13 | Viewed by 3398
Abstract
Bone metastasis is the terminal stage disease of prostate, breast, renal, and lung cancers, and currently no therapeutic approach effectively cures or prevents its progression to bone metastasis. One of the hurdles to the development of new drugs for bone metastasis is the [...] Read more.
Bone metastasis is the terminal stage disease of prostate, breast, renal, and lung cancers, and currently no therapeutic approach effectively cures or prevents its progression to bone metastasis. One of the hurdles to the development of new drugs for bone metastasis is the complexity and heterogeneity of the cellular components in the metastatic bone microenvironment. For example, bone cells, including osteoblasts, osteoclasts, and osteocytes, and the bone marrow cells of diverse hematopoietic lineages interact with each other via numerous cytokines and receptors. c-Met tyrosine kinase receptor and its sole ligand hepatocyte growth factor (HGF) are enriched in the bone microenvironment, and their expression correlates with the progression of bone metastasis. However, no drugs or antibodies targeting the c-Met/HGF signaling axis are currently available in bone metastatic patients. This significant discrepancy should be overcome by further investigation of the roles and regulation of c-Met and HGF in the metastatic bone microenvironment. This review paper summarizes the key findings of c-Met and HGF in the development of novel therapeutic approaches for bone metastasis. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
17 pages, 1576 KiB  
Review
Hepatocyte Growth Factor: A Microenvironmental Resource for Leukemic Cell Growth
by Paolo Giannoni, Franco Fais, Giovanna Cutrona and Daniela de Totero
Int. J. Mol. Sci. 2019, 20(2), 292; https://doi.org/10.3390/ijms20020292 - 12 Jan 2019
Cited by 8 | Viewed by 3998
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the progressive expansion of B lymphocytes CD5+/CD23+ in peripheral blood, lymph-nodes, and bone marrow. The pivotal role played by the microenvironment in disease pathogenesis has become increasingly clear. We demonstrated that bone marrow stromal cells and [...] Read more.
Chronic lymphocytic leukemia (CLL) is characterized by the progressive expansion of B lymphocytes CD5+/CD23+ in peripheral blood, lymph-nodes, and bone marrow. The pivotal role played by the microenvironment in disease pathogenesis has become increasingly clear. We demonstrated that bone marrow stromal cells and trabecular bone cells sustain survival of leukemic B cells through the production of hepatocyte growth factor (HGF). Indeed the trans-membrane kinase receptor for HGF, c-MET, is expressed on CLL cells and STAT3 TYR705 or AKT phosphorylation is induced after HGF/c-MET interaction. We have further observed that c-MET is also highly expressed in a peculiar type of cells of the CLL-microenvironment showing nurturing features for the leukemic clone (nurse-like cells: NLCs). Since HGF treatment drives monocytes toward the M2 phenotype and NLCs exhibit features of tumor associated macrophages of type 2 we suggested that HGF, released either by cells of the microenvironment or leukemic cells, exerts a double effect: i) enhances CLL cells survival and ii) drives differentiation of monocytes-macrophages to an oriented immune suppressive phenotype. We here discuss how paracrine, but also autocrine production of HGF by malignant cells, may favor leukemic clone expansion and resistance to conventional drug treatments in CLL, as well as in other hematological malignancies. Novel therapeutic approaches aimed to block HGF/c-MET interactions are further proposed. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Figure 1

17 pages, 2601 KiB  
Review
MET/HGF Co-Targeting in Pancreatic Cancer: A Tool to Provide Insight into the Tumor/Stroma Crosstalk
by Chiara Modica, Dora Tortarolo, Paolo M. Comoglio, Cristina Basilico and Elisa Vigna
Int. J. Mol. Sci. 2018, 19(12), 3920; https://doi.org/10.3390/ijms19123920 - 07 Dec 2018
Cited by 19 | Viewed by 4608
Abstract
The ‘onco-receptor’ MET (Hepatocyte Growth Factor Receptor) is involved in the activation of the invasive growth program that is essential during embryonic development and critical for wound healing and organ regeneration during adult life. When aberrantly activated, MET and its stroma-secreted ligand HGF [...] Read more.
The ‘onco-receptor’ MET (Hepatocyte Growth Factor Receptor) is involved in the activation of the invasive growth program that is essential during embryonic development and critical for wound healing and organ regeneration during adult life. When aberrantly activated, MET and its stroma-secreted ligand HGF (Hepatocyte Growth Factor) concur to tumor onset, progression, and metastasis in solid tumors, thus representing a relevant target for cancer precision medicine. In the vast majority of tumors, wild-type MET behaves as a ‘stress-response’ gene, and relies on ligand stimulation to sustain cancer cell ‘scattering’, invasion, and protection form apoptosis. Moreover, the MET/HGF axis is involved in the crosstalk between cancer cells and the surrounding microenvironment. Pancreatic cancer (namely, pancreatic ductal adenocarcinoma, PDAC) is an aggressive malignancy characterized by an abundant stromal compartment that is associated with early metastases and resistance to conventional and targeted therapies. Here, we discuss the role of the MET/HGF axis in tumor progression and dissemination considering as a model pancreatic cancer, and provide a proof of concept for the application of dual MET/HGF inhibition as an adjuvant therapy in pancreatic cancer patients. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Figure 1

17 pages, 852 KiB  
Review
HGF/c-MET Signaling in Melanocytes and Melanoma
by Malgorzata Czyz
Int. J. Mol. Sci. 2018, 19(12), 3844; https://doi.org/10.3390/ijms19123844 - 03 Dec 2018
Cited by 70 | Viewed by 7806
Abstract
Hepatocyte growth factor (HGF)/ mesenchymal-epithelial transition factor (c-MET) signaling is involved in complex cellular programs that are important for embryonic development and tissue regeneration, but its activity is also utilized by cancer cells during tumor progression. HGF and c-MET usually mediate heterotypic cell–cell [...] Read more.
Hepatocyte growth factor (HGF)/ mesenchymal-epithelial transition factor (c-MET) signaling is involved in complex cellular programs that are important for embryonic development and tissue regeneration, but its activity is also utilized by cancer cells during tumor progression. HGF and c-MET usually mediate heterotypic cell–cell interactions, such as epithelial–mesenchymal, including tumor–stroma interactions. In the skin, dermal fibroblasts are the main source of HGF. The presence of c-MET on keratinocytes is crucial for wound healing in the skin. HGF is not released by normal melanocytes, but as melanocytes express c-MET, they are receptive to HGF, which protects them from apoptosis and stimulates their proliferation and motility. Dissimilar to melanocytes, melanoma cells not only express c-MET, but also release HGF, thus activating c-MET in an autocrine manner. Stimulation of the HGF/c-MET pathways contributes to several processes that are crucial for melanoma development, such as proliferation, survival, motility, and invasiveness, including distant metastatic niche formation. HGF might be a factor in the innate and acquired resistance of melanoma to oncoprotein-targeted drugs. It is not entirely clear whether elevated serum HGF level is associated with low progression-free survival and overall survival after treatment with targeted therapies. This review focuses on the role of HGF/c-MET signaling in melanoma with some introductory information on its function in skin and melanocytes. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Graphical abstract

13 pages, 595 KiB  
Review
HGF/MET and the Immune System: Relevance for Cancer Immunotherapy
by Federica Papaccio, Carminia Maria Della Corte, Giuseppe Viscardi, Raimondo Di Liello, Giovanna Esposito, Francesca Sparano, Fortunato Ciardiello and Floriana Morgillo
Int. J. Mol. Sci. 2018, 19(11), 3595; https://doi.org/10.3390/ijms19113595 - 14 Nov 2018
Cited by 71 | Viewed by 5931
Abstract
An overactivation of hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (MET) axis promotes tumorigenesis and tumor progression in various cancer types. Research data recently evidenced that HGF/MET signaling is also involved also in the immune response, mainly modulating dendritic cells functions. In general, the [...] Read more.
An overactivation of hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (MET) axis promotes tumorigenesis and tumor progression in various cancer types. Research data recently evidenced that HGF/MET signaling is also involved also in the immune response, mainly modulating dendritic cells functions. In general, the pathway seems to play an immunosuppressive role, thus hypothesizing that it could constitute a mechanism of primary and acquired resistance to cancer immunotherapy. Recently, some approaches are being developed, including drug design and cell therapy to combine MET and programmed cell death receptor-1 (PD-1)/programmed cell death receptor-ligand 1 (PD-L1) inhibition. This approach could represent a new weapon in cancer therapy in the future. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Graphical abstract

11 pages, 1408 KiB  
Review
Hepatocyte Growth Factor Activator: A Proteinase Linking Tissue Injury with Repair
by Tsuyoshi Fukushima, Shuichiro Uchiyama, Hiroyuki Tanaka and Hiroaki Kataoka
Int. J. Mol. Sci. 2018, 19(11), 3435; https://doi.org/10.3390/ijms19113435 - 01 Nov 2018
Cited by 56 | Viewed by 5695
Abstract
Hepatocyte growth factor (HGF) promotes pleiotropic signaling through its specific receptor tyrosine kinase, MET. As such, it has important roles in the regeneration of injured tissues. Since HGF is produced mainly by mesenchymal cells and MET is expressed in most epithelial, endothelial and [...] Read more.
Hepatocyte growth factor (HGF) promotes pleiotropic signaling through its specific receptor tyrosine kinase, MET. As such, it has important roles in the regeneration of injured tissues. Since HGF is produced mainly by mesenchymal cells and MET is expressed in most epithelial, endothelial and somatic stem cells, HGF functions as a typical paracrine growth factor. HGF is secreted as an inactive precursor (proHGF) and requires proteolytic activation to initiate HGF-induced MET signaling. HGF activator (HGFAC) is a serum activator of proHGF and produces robust HGF activities in injured tissues. HGFAC is a coagulation factor XII-like serine endopeptidase that circulates in the plasma as a zymogen (proHGFAC). Thrombin, kallikrein-related peptidase (KLK)-4 or KLK-5 efficiently activates proHGFAC. The activated HGFAC cleaves proHGF at Arg494-Val495, resulting in the formation of the active disulfide-linked heterodimer HGF. Macrophage stimulating protein, a ligand of RON, is also activated by HGFAC in vivo. Although HGFAC functions primarily at the site of damaged tissue, a recent report has suggested that activated HGFAC relays a signal to stem cells in non-injured tissues via proHGF activation in the stem cell niche. This review focuses on current knowledge regarding HGFAC-mediated proHGF activation and its roles in tissue regeneration and repair. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Figure 1

13 pages, 956 KiB  
Review
Extracellular Matrix Influencing HGF/c-MET Signaling Pathway: Impact on Cancer Progression
by Heydi Noriega-Guerra and Vanessa Morais Freitas
Int. J. Mol. Sci. 2018, 19(11), 3300; https://doi.org/10.3390/ijms19113300 - 24 Oct 2018
Cited by 50 | Viewed by 5157
Abstract
The extracellular matrix (ECM) is a crucial component of the tumor microenvironment involved in numerous cellular processes that contribute to cancer progression. It is acknowledged that tumor–stromal cell communication is driven by a complex and dynamic network of cytokines, growth factors and proteases. [...] Read more.
The extracellular matrix (ECM) is a crucial component of the tumor microenvironment involved in numerous cellular processes that contribute to cancer progression. It is acknowledged that tumor–stromal cell communication is driven by a complex and dynamic network of cytokines, growth factors and proteases. Thus, the ECM works as a reservoir for bioactive molecules that modulate tumor cell behavior. The hepatocyte growth factor (HGF) produced by tumor and stromal cells acts as a multifunctional cytokine and activates the c-MET receptor, which is expressed in different tumor cell types. The HGF/c-MET signaling pathway is associated with several cellular processes, such as proliferation, survival, motility, angiogenesis, invasion and metastasis. Moreover, c-MET activation can be promoted by several ECM components, including proteoglycans and glycoproteins that act as bridging molecules and/or signal co-receptors. In contrast, c-MET activation can be inhibited by proteoglycans, matricellular proteins and/or proteases that bind and sequester HGF away from the cell surface. Therefore, understanding the effects of ECM components on HGF and c-MET may provide opportunities for novel therapeutic strategies. Here, we give a short overview of how certain ECM components regulate the distribution and activation of HGF and c-MET. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Figure 1

17 pages, 4237 KiB  
Review
HGF/c-MET: A Promising Therapeutic Target in the Digestive System Cancers
by Hongli Zhang, Qingqing Feng, Wei-Dong Chen and Yan-Dong Wang
Int. J. Mol. Sci. 2018, 19(11), 3295; https://doi.org/10.3390/ijms19113295 - 23 Oct 2018
Cited by 34 | Viewed by 6252
Abstract
The HGF/c-MET pathway is active in the development of digestive system cancers, indicating that inhibition of HGF/c-MET signaling may have therapeutic potential. Various HGF/c-MET signaling inhibitors, mainly c-MET inhibitors, have been tested in clinical trials. The observed efficacy and adverse events of some [...] Read more.
The HGF/c-MET pathway is active in the development of digestive system cancers, indicating that inhibition of HGF/c-MET signaling may have therapeutic potential. Various HGF/c-MET signaling inhibitors, mainly c-MET inhibitors, have been tested in clinical trials. The observed efficacy and adverse events of some c-MET inhibitors were not very suitable for treating digestive system cancers. The development of new HGF/c-MET inhibitors in preclinical studies may bring promising treatments and synergistic combination (traditional anticancer drugs and c-MET inhibitors) strategies provided anacceptable safety and tolerability. Insights into miRNA biology and miRNA therapeutics have made miRNAs attractive tools to inhibit HGF/c-MET signaling. Recent reports show that several microRNAs participate in inhibiting HGF/c-MET signaling networks through antagonizing c-MET or HGF in digestive system cancers, and the miRNAs-HGF/c-MET axis plays crucial and novel roles for cancer treatment. In the current review, we will discuss recent findings about inhibitors of HGF/c-MET signaling in treating digestive system cancers, and how miRNAs regulate digestive system cancers via mediating HGF/c-MET pathway. Full article
(This article belongs to the Special Issue Hepatocyte Growth Factor (HGF))
Show Figures

Graphical abstract

Back to TopTop