scispace - formally typeset
Search or ask a question

Showing papers on "Hepatocyte growth factor published in 1996"


Journal ArticleDOI
TL;DR: While HGF was originally identified as a potent mitogen for mature hepatocytes, the biological functions of this factor reach far beyond the original identifications and use of HGF for purposes of therapeutics is being given increasing attention.
Abstract: Specific tissue interactions between epithelia and mesenchyme (or stroma), e.g., epithelial-mesenchymal (or -stromal) interactions mediate crucial aspects of normal development and tissue regeneration. These events affect tissue induction, organogenesis, cell movement, and morphogenesis of multicellular structures. Extensive and diverse studies have established that hepatocyte growth factor (HGF), a ligand for the c-met protooncogene product of receptor tyrosine kinase, is a mesenchymal- or stromal-derived multipotent polypeptide which mediates epithelial-mesenchymal interactions. During embryogenesis, HGF supports organogenesis and morphogenesis of various tissues and organs, including the liver, kidney, lung, gut, mammary gland, tooth, skeletal system, etc. In adult tissues, HGF elicits a potent organotrophic function which supports regeneration of organs including the liver, kidney, and lung. In the brain, HGF is a new member of the family of neurotrophic factors. In neoplastic tissue, HGF is involved in tumor invasion and metastasis, through tumor-stromal interactions. While HGF was originally identified as a potent mitogen for mature hepatocytes, the biological functions of this factor reach far beyond the original identifications. Such being the case, use of HGF for purposes of therapeutics is being given increasing attention.

656 citations


Journal ArticleDOI
TL;DR: The hypothesis that mature hepatocytes can function as or be a source of bipotential facultative hepatic stem cells (hepatoblasts) is supported and evidence for the growth factor and matrix signals that govern these complex phenotypic transitions of facultative stem cells are provided.
Abstract: Mature adult parenchymal hepatocytes, typically of restricted capacity to proliferate in culture, can now enter into clonal growth under the influence of hepatocyte growth factor (scatter factor) (HGF/SF), epidermal growth factor (EGF), and transforming growth factor alpha (TGFalpha) in the presence of a new chemically defined medium (HGM). The expanding populations of hepatocytes lose expression of hepatocyte specific genes (albumin, cytochrome P450 IIB1), acquire expression of markers expressed by bile duct epithelium (cytokeratin 19), produce TGFalpha and acidic FGF and assume a very simplified morphologic phenotype by electron microscopy. A major change associated with this transition is the decrease in ratio between transcription factors C/EBPalpha and C/EBPbeta, as well as the emergence in the proliferating hepatocytes of transcription factors AP1, NFkappaB. The liver associated transcription factors HNFI, HNF3, and HNF4 are preserved throughout this process. After population expansion and clonal growth, the proliferating hepatocytes can return to mature hepatocyte phenotype in the presence of EHS gel (Matrigel). This includes complete restoration of electron microscopic structure and albumin expression. The hepatocyte cultures however can instead be induced to form acinar/ductular structures akin to bile ductules (in the presence of HGF/SF and type I collagen). These transformations affect the entire population of the hepatocytes and occur even when DNA synthesis is inhibited. Similar acinar/ductular structures are seen in embryonic liver when HGF/SF and its receptor are expressed at high levels. These findings strongly support the hypothesis that mature hepatocytes can function as or be a source of bipotential facultative hepatic stem cells (hepatoblasts). These studies also provide evidence for the growth factor and matrix signals that govern these complex phenotypic transitions of facultative stem cells which are crucial for recovery from acute and chronic liver injury.

514 citations


Journal ArticleDOI
01 Dec 1996-Neuron
TL;DR: Results indicate that, in addition to functioning as a mitogen, a motogen, and a morphogen in nonneural systems, HGF/SF can function as a guidance and survival factor in the developing nervous system.

409 citations


Journal ArticleDOI
TL;DR: It is shown that c-met expression is markedly reduced in the lateral dermomyotome of Splotch embryos lacking Pax3, and a potential Pax3 binding site in the human c-MET promoter that may contribute to direct transcriptional regulation is identified.
Abstract: Pax3 is a transcription factor whose expression has been used as a marker of myogenic precursor cells arising in the lateral somite destined to migrate to and populate the limb musculature. Accruing evidence indicates that the embryologic origins of axial and appendicular muscles are distinct, and limb muscle abnormalities in both mice and humans harboring Pax3 mutations support this distinction. The mechanisms by which Pax3 affects limb muscle development are unknown. The tyrosine kinase receptor for hepatocyte growth factor/scatter factor encoded by the c-met protooncogene is also expressed in limb muscle progenitors and, like Pax-3, is required in the mouse for limb muscle development. Here, we show that c-met expression is markedly reduced in the lateral dermomyotome of Splotch embryos lacking Pax3. We show that Pax3 can stimulate c-met expression in cultured cells, and we identify a potential Pax3 binding site in the human c-MET promoter that may contribute to direct transcriptional regulation. In addition, we have found that several cell lines derived from patients with rhabdomyosarcomas caused by a t(2;13) chromosomal translocation activating PAX3 express c-MET, whereas those rhabdomyosarcoma cell lines examined without the translocation do not. These results are consistent with a model in which Pax3 modulates c-met expression in the lateral dermomyotome, a function that is required for the appropriate migration of these myogenic precursors to the limb where the ligand for c-met (hepatocyte growth factor/scatter factor) is expressed at high levels.

380 citations


Journal ArticleDOI
TL;DR: It is shown that HGF/SF-Met signalling in the human leiomyosarcoma cell line SK-LMS-1 enhances its in vivo tumorigenicity, an effect for which the mitogenicity of this signalling pathway is likely to play a role.
Abstract: Hepatocyte growth factor/scatter factor (HGF/SF) is a pleiotropic effector of cells expressing the Met tyrosine kinase receptor. Although HGF/SF is synthesized by mesenchymal cells and acts predominantly on epithelial cells, we have recently demonstrated that human sarcoma cell lines often inappropriately express high levels of Met and respond mitogenically to HGF/SF. In the present report we show that HGF/SF-Met signalling in the human leiomyosarcoma cell line SK-LMS-1 enhances its in vivo tumorigenicity, an effect for which the mitogenicity of this signalling pathway is likely to play a role. In addition, we found that HGF/SF-Met signalling dramatically induces the in vitro invasiveness and in vivo metastatic potential of these cells. We have studied the molecular basis by which HGFSF-Met signalling mediates the invasive phenotype. A strong correlation has previously been demonstrated between the activation of the urokinase plasminogen activator (uPA) proteolysis network and the acquisition of the invasive-metastatic phenotype, and we show here that HGF/SF-Met signalling significantly increases the protein levels of both uPA and its cellular receptor in SK-LMS-1 cells. This results in elevated levels of cell-associated uPA and enhanced plasmin-generating ability by these cells. These studies couple HGF/SF-Met signalling to the activation of proteases that mediate dissolution of the extracellular matrix-basement membrane, and important property for cellular invasion-metastasis.

325 citations


Journal ArticleDOI
TL;DR: The finding of a link between growth factor receptors and the anti‐apoptotic machinery fills a gap in the understanding of the molecular events regulating programmed cell death.
Abstract: The mechanisms by which apoptosis is prevented by survival factors are largely unknown. Using an interaction cloning approach, we identified a protein that binds to the intracellular domain of the hepatocyte growth factor (HGF) receptor. This protein was identified as BAG-1, a recently characterized Bcl-2 functional partner, which prolongs cell survival through unknown mechanisms. Overexpression of BAG-1 in liver progenitor cells enhances protection from apoptosis by HGF. Association of the receptor with BAG-1 occurs in intact cells, is mediated by the C-terminal region of BAG-1 and is independent from tyrosine phosphorylation of the receptor. Formation of the complex is increased rapidly following induction of apoptosis. BAG-1 also enhances platelet-derived growth factor (PDGF)-mediated protection from apoptosis and associates with the PDGF receptor. Microinjection or transient expression of BAG-1 deletion mutants shows that both the N- and the C-terminal domains are required for protection from apoptosis. The finding of a link between growth factor receptors and the anti-apoptotic machinery fills a gap in the understanding of the molecular events regulating programmed cell death.

320 citations


Journal ArticleDOI
01 Nov 1996-Cell
TL;DR: The data show that the requirements for Met signaling vary depending on the tissue and reveal a novel role for HGF/ Met in late myogenesis, as well as highlighting the importance of knowing the carrier and removal status of canine coronavirus.

313 citations


Journal ArticleDOI
TL;DR: Several model systems developed to investigate the role of HGF/SF-Met signaling in malignancy are discussed and additional data regarding the expression of these molecules in human tumors are described.
Abstract: Hepatocyte growth factor/scatter factor (HGF/SF) is a pleiotropic effector of cells expressing the Met tyrosine kinase receptor. While HGF/SF-Met signaling clearly plays a role in a variety of normal cellular processes, this signaling pathway has also been implicated in the generation and metastatic spread of tumors. This review discusses in detail several model systems that have been developed to investigate the role of HGF/SF-Met signaling in malignancy and describes additional data regarding the expression of these molecules in human tumors. Collectively the findings support a role for this receptor-ligand pair in human malignancy.

268 citations


Journal ArticleDOI
TL;DR: A critical summary of the known effects of growth factors on skeletal muscle regeneration is presented, showing TGF beta has a major influence on the reorganisation of the extracellular matrix.

260 citations


Journal Article
TL;DR: The finding that HGFR is expressed by both benign and malignant epithelium, and its not restricted to duct-forming structures, suggests that, although the potential for HGF/HGFR binding is maintained in malignancy, the response to ligand binding at the level of the receptor or the cellular response to receptor activation may change at some point during progression.
Abstract: Expression of hepatocyte growth factor (HGF) and HGF receptor (HGFR, product of the met proto-oncogene) mRNA were examined by nonisotopic in situ hybridization in a spectrum of benign and malignant human breast tissues. mRNA for both HGFR and HGF was detected in benign ductal epithelium. Epithelial expression of HGF mRNA was particularly intense in regions of ductal epithelial hyperplasia. Positive expression of HGF (but not HGFR) mRNA was also found in adipocytes, endothelial cells, and to varying degrees in stromal fibroblasts. In 12 of 12 cases of ductal carcinoma in situ and infiltrating ductal carcinoma, carcinoma cells showed a heterogeneous pattern of expression for both HGFR and HGF mRNA. In infiltrating ductal carcinomas, intense expression of HGFR mRNA was not restricted to ductular structures but as also seen in non-duct-forming carcinoma cells. The same zones of the tumors (most commonly at the advancing margins) that expressed strongly HGFR mRNA often were also strongly positive for HGF mRNA, suggesting a possible autocrine effect. The expression pattern of HGFR protein in 25 cases including the same series of tissues used for in situ hybridization analysis was similar to that of HGFR mRNA, as determined by an immunoperoxidase technique. The finding that HGFR is expressed by both benign and malignant epithelium, and its not restricted to duct-forming structures, suggests that, although the potential for HGF/HGFR binding is maintained in malignancy, the response to ligand binding at the level of the receptor or the cellular response to receptor activation may change at some point during progression.

255 citations


Journal ArticleDOI
TL;DR: It is found that the HGF-converting activity in the injured liver was inhibited by an anti-HGFA antibody, and it appears likely that HGFA is the key enzyme that regulates the activity of HGF in thejured tissues.

Patent
31 May 1996
TL;DR: Hepatocyte growth factor (HGF) receptor antagonists include HGF receptor antibodies and fragments thereof as mentioned in this paper, which can be employed to block binding of HGF to HGF receptors or substantially inhibit HGF activation.
Abstract: Hepatocyte growth factor (HGF) receptor antagonists are provided. The HGF receptor antagonists include HGF receptor antibodies and fragments thereof. The HGF receptor antagonists can be employed to block binding of HGF to HGF receptors or substantially inhibit HGF receptor activation. The HGF receptor antagonists may be included in pharmaceutical compositions, articles of manufacture, or kits. Methods of treating cancer using the HGF receptor antagonists are also provided.

Journal ArticleDOI
15 Nov 1996-Blood
TL;DR: The mean HGF level in serum of myeloma patients at diagnosis was more than fourfold higher than the mean level in normal controls, and possible implications of HGF/c-met expression for the pathophysiology of multipleMyeloma are discussed.

Journal ArticleDOI
TL;DR: Data show that HGF/SF is overexpressed and consistently activated in non-small-cell lung carcinomas and may contribute to the invasive growth of lung cancer.
Abstract: Hepatocyte growth factor/scatter factor (HGF/SF) stimulates the invasive growth of epithelial cells via the c-MET oncogene-encoded receptor. In normal lung, both the receptor and the ligand are detected, and the latter is known to be a mitogenic and a motogenic factor for both cultured bronchial epithelial cells and non-small-cell carcinoma lines. Here, ligand and receptor expression was examined in 42 samples of primary human non-small-cell lung carcinoma of different histotype. Each carcinoma sample was compared with adjacent normal lung tissue. The Met/HGF receptor was found to be 2 to 10-fold increased in 25% of carcinoma samples (P = 0.0113). The ligand, HGF/SF, was found to be 10 to 100-fold overexpressed in carcinoma samples (P < 0.0001). Notably, while HGF/SF was occasionally detectable and found exclusively as a single-chain inactive precursor in normal tissues, it was constantly in the biologically-active heterodimeric form in carcinomas. Immunohistochemical staining showed homogeneous expression of both the receptor and the ligand in carcinoma samples, whereas staining was barely detectable in their normal counterparts. These data show that HGF/SF is overexpressed and consistently activated in non-small-cell lung carcinomas and may contribute to the invasive growth of lung cancer.

Journal ArticleDOI
15 Mar 1996-Cancer
TL;DR: The quantitation of endothelial growth factors is thought to be useful to assess angiogenic phenotype in the tumor to promote the neovascularization in human breast cancer.
Abstract: BACKGROUND Recent clinical studies have demonstrated that tumor angiogenesis is a potent prognostic indicator for breast cancer patients. The quantitation of endothelial growth factors is thought to be useful to assess angiogenic phenotype in the tumor. Among the many new endothelial growth factors, vascular endothelial growth factor (VEGF) is known to be particularly responsible for promoting the neovascularization in human breast cancer. METHODS Intratumoral protein levels of VEGF were measured by enzymatic immunoassay in 135 primary breast cancer tissues. The VEGF levels were compared with the microvessel density evaluated by immunostaining the endothelial antigen and also were compared with intratumoral protein levels of other endothelial growth factors, including basic fibroblast growth factor (bFGF) and hepatocyte growth factor (HGF). RESULTS Intratumoral VEGF concentrations varied from 3.3 pg/mg protein to 2032 pg/mg protein (average 148 pg/mg protein). An immunocytochemical analysis using anti-VEGF antibody confirmed that VEGF was located mainly in the cytoplasm of the tumor cells. The VEGF concentrations were significantly higher in vascularly rich tumors than in vascularly poor tumors. No significant association was found between VEGF concentrations and the two other endothelial growth factor concentrations. CONCLUSIONS The quantitation of intratumoral VEGF levels seems to be useful for assessing the activity of tumor angiogenesis. Cancer 1996;77:1101-6.

Journal ArticleDOI
TL;DR: It is shown that the HGF receptor is expressed by human primary osteoclasts, by osteoclast-like cell lines, and by osteoblasts, suggesting the possibility of an autocrine regulation of the osteoplast by HGF and a paracrine regulation ofThe osteoblast by the H GF produced by the osteOClast.
Abstract: Hepatocyte growth factor (HGF), also known as scatter factor, is a powerful motogen, mitogen, and morphogen produced by cells of mesodermal origin, acting on epithelial and endothelial cells. Its receptor is the tyrosine kinase encoded by the c-MET protooncogene. We show that the HGF receptor is expressed by human primary osteoclasts, by osteoclast-like cell lines, and by osteoblasts. In both cell lineages, HGF stimulation triggers the receptor kinase activity and autophosphorylation. In osteoclasts, HGF receptor activation is followed by increase in intracellular Ca2+ concentration and by activation of the pp60c-Src kinase. HGF induces changes in osteoclast shape and stimulates chemotactic migration and DNA replication. Osteoblasts respond to HGF by entering the cell cycle, as indicated by stimulation of DNA synthesis. Interestingly, osteoclasts were found to synthesize and secrete biologically active HGF. These data strongly suggest the possibility of an autocrine regulation of the osteoclast by HGF and a paracrine regulation of the osteoblast by the HGF produced by the osteoclast.

Journal ArticleDOI
TL;DR: The results suggest that HGF from microglia has neurotrophic activity on the CNS neurons and plays significant roles in the development of the CNS.
Abstract: Although the expression of hepatocyte growth factor (HGF) and its receptor, proto-oncogene c-met, has been demonstrated in the central nervous system (CNS), the function of HGF in the CNS was not fully understood. In the present studies, we determined the effects of HGF on neuronal development in neocortical explant and mesencephalic neurons obtained from embryonic rat brain. HGF clearly enhanced neurite outgrowth in neocortical explants. In the mesencephalic culture, the number of tyrosine hydroxylase (TH)-positive neurons was significantly higher in the HGF-treated wells and the neurites of the TH-positive neurons appear to be more developed. Moreover, the dopamine uptake into mesencephalic neurons was also enhanced by HGF treatment, indicating that HGF promotes the survival and/or maturation of mesencephalic dopaminergic neurons. In both neocortical explants and mesencephalic neurons, c-met autophosphorylation was induced by HGF and MAP kinase activation was also detected in the neocortical explant. Furthermore, Western blot analysis of the cultured CNS cells revealed that HGF was expressed mainly in microglia. These results suggest that HGF from microglia has neurotrophic activity on the CNS neurons and plays significant roles in the development of the CNS. © 1996 Wiley-Liss, Inc.

Journal ArticleDOI
TL;DR: HGF can exert stimulating effects on endothelial cell growth, but not on VSMC growth, in an additive manner with bFGF but not with VEGF, demonstrating that HGF may provide the opportunity for a new therapeutic strategy for vascular diseases in which the abnormalities are vasoconstriction and pathological growth.
Abstract: OBJECTIVE To seek an endothelium-specific growth factor by examining the mitogenic effects of hepatocyte growth factor (HGF) on endothelial cells and on vascular smooth muscle cells (VSMC). METHODS Rat and human endothelial cells and VSMC were employed. DNA, RNA and protein synthesis were measured by using [3H]-thymidine, uridine and leucine. Coculture of endothelial cells with VSMC was also performed to study the role of endothelial cells. RESULTS Coculture of endothelial cells with VSMC resulted in a significant decrease in DNA synthesis of VSMC. HGF, as well as basic fibroblast growth factor (bFGF), stimulated DNA, RNA and protein synthesis by endothelial cells in a dose-dependent manner. Interestingly, co-incubation of endothelial cells with HGF and bFGF resulted in an additive stimulation of DNA synthesis. Similarly, HGF and interleukin-1 alpha and -6 stimulated DNA synthesis by coronary endothelial cells, whereas interleukin-1 beta and transforming growth factor-beta (TGF-beta) did not. However, HGF showed markedly different actions from bFGF on VSMC growth. bFGF, TGF-beta, interleukin-1 alpha, -1 beta and -6 stimulated DNA synthesis in VSMC significantly, whereas HGF did not. Finally, we examined the mitogenic effect of HGF on human aortic endothelial cells and VSMC. Incubation with HGF increased DNA synthesis and growth by endothelial cells in a dose-dependent manner, whose degree was significantly greater than those with bFGF, vascular endothelial growth factor (VEGF) and interleukin-6. Addition of HGF and VEGF showed no additive effect on DNA synthesis in endothelial cells, in contrast to those of bFGF and HGF. On the other hand, bFGF, but not HGF and VEGF, stimulated DNA synthesis in VSMC. CONCLUSION These results demonstrate that HGF can exert stimulating effects on endothelial cell growth, but not on VSMC growth, in an additive manner with bFGF but not with VEGF. These characteristics of HGF as an endothelium-specific growth factor may provide the opportunity for a new therapeutic strategy for vascular diseases in which the abnormalities were vasoconstriction and pathological growth.

Journal ArticleDOI
TL;DR: Functional evidence is provided that inappropriate expression of SF/HGF in transgenic mice influences the development of two distinct migratory cell lineages, resulting in ectopic skeletal muscle formation and melanosis in the central nervous system, and patterned hyperpigmentation of the skin.
Abstract: Factors that regulate cellular migration during embryonic development are essential for tissue and organ morphogenesis. Scatter factor/hepatocyte growth factor (SF/HGF) can stimulate motogenic and morphogenetic activities in cultured epithelial cells expressing the Met tyrosine kinase receptor and is essential for development; however, the precise physiological role of SF/HGF is incompletely understood. Here we provide functional evidence that inappropriate expression of SF/HGF in transgenic mice influences the development of two distinct migratory cell lineages, resulting in ectopic skeletal muscle formation and melanosis in the central nervous system, and patterned hyperpigmentation of the skin. Committed TRP-2 positive melanoblasts were found to be situated aberrantly within defined regions of the transgenic embryo, including the neural tube, which overproduced SF/RGF. Our data strongly suggest that SF/HGF possesses physiologically relevant scatter activity, and functions as a true morphogenetic factor by regulating migration and/or differentiation of select populations of premyogenic and neural crest cells during normal mammalian embryogenesis.

Journal ArticleDOI
TL;DR: Definition of the mechanisms by which primary mitogens stimulate liver cell proliferation may elucidate the nature of the signals responsible for triggering the entry into cell cycle.
Abstract: Liver cell growth can be induced in two distinct patterns: compensatory regeneration and direct hyperplasia. In the former, DNA synthesis is preceded by a loss of liver cells such as seen after partial resection of the liver or cell necrosis, whereas in direct hyperplasia, DNA synthesis is stimulated without cell loss. During the past decade, considerable advances have been made in understanding molecular mechanisms of the compensatory regeneration. There is increasing evidence that hepatocyte proliferation induced by some primary mitogens is mediated by patterns of growth factor modulation and signal transduction different from those of compensatory regeneration. Indeed, whereas activation of transcription factors such as NF-kappa B and increased expression of immediate early genes such as c-fos, c-jun, egr-1, and c-myc are induced during compensatory regeneration, such changes are not observed during hyperplasia induced by certain primary mitogens. In addition, although experimental evidence suggests a critical role for growth factors such as hepatocyte growth factor and transforming growth factor-alpha for the progression into cell cycle of competent hepatocytes in compensatory regeneration, these growth factors do not appear to play a major role in direct hyperplasia. One class of primary mitogens may trigger their actions through tumor necrosis factor-alpha, and the other by activation of nuclear hormone receptors. The differences in molecular events observed between liver regeneration and direct hyperplasia may affect differently the initiation step of chemical hepatocarcinogenesis. Whereas the former supports initiation by chemicals, the latter does not. A similar lack of effect on promotion of carcinogen-altered cells has also been observed after acute treatment with some primary mitogens. Definition of the mechanisms by which primary mitogens stimulate liver cell proliferation may elucidate the nature of the signals responsible for triggering the entry into cell cycle. Furthermore, due to their low toxicity, primary liver mitogens could have significant clinical applications in gene transfer and liver transplantation.

Journal ArticleDOI
01 Sep 1996-Diabetes
TL;DR: These are the first experiments to describe the interactions of specific matrices and growth factors in the ex vivo expansion of human fetal pancreatic cells, and they show the importance of cell aggregates in the context of cellular and molecular events that might positively influence islet cell transplantation.
Abstract: Ex vivo expansion of human fetal pancreatic endocrine cells is important for biological studies and as a potential tissue source for transplantation in insulin-deficient states. In tissue culture experiments involving the use of hepatocyte growth factor/scatter factor and selected extracellular matrices, we obtained a 30-fold increase in cell number of human fetal pancreatic epithelial cells. This proliferation in monolayer culture was associated with marked downregulation of insulin and glucagon gene expression. However, gene expression increased when the cells were combined into three-dimensional aggregates, suggesting that cell-cell contact mediated mechanisms regulate the transcription of islet-specific genes, a process enhanced by nicotinamide (NIC). After transplantation into nude mice, either as cell suspensions or aggregates, only the cell aggregates treated with NIC developed into mature functional islet-like structures. These are the first experiments to describe the interactions of specific matrices and growth factors in the ex vivo expansion of human fetal pancreatic cells, and they also show the importance of cell aggregates in the context of cellular and molecular events that might positively influence islet cell transplantation.

Journal ArticleDOI
TL;DR: Results indicate that HGF reduces the acinar cell-like property of AR42J cells and converts them into insulin-producing cells and the effect of HGF was markedly enhanced by activin A.
Abstract: Pancreatic AR42J cells are derived from acinar cells and express both exocrine and neuroendocrine properties. We have recently shown that these cells convert into insulin-producing cells in vitro after treatment with activin A and betacellulin. Here, we investigated the effect of hepatocyte growth factor (HGF) in those cells. When AR42J cells were incubated with HGF, DNA synthesis was attenuated, and the amylase content was reduced in a concentration-dependent manner. HGF-treated cells extended processes, but bundle formation was not observed using an antibody against tubulin. Reverse both insulin and pancreatic polypeptide (PP) were expressed in HGF-treated, but not naive, AR42J cells. Immunocytochemical analysis indicated that approximately 3% of the HGF-treated cells were stained with antiinsulin antibody, and some were also stained with anti-PP antibody. When AR42J cells were exposed to a combination of activin A and HGF, cells extended longer processes, and over 10% of them were stained with antiinsulin antibody. In these cells, messenger RNAs for insulin, PP, glucose transporter 2, and glucokinase, but not those for glucagon or somatostatin, were expressed. A subclone of AR42J cells, AR42J-B13, was obtained. Most of the AR42J-B13 cells converted to insulin-producing cells after the incubation with activin A and HGF. Insulin secretion was augmented by tolbutamide, depolarizing concentrations of potassium, carbachol, and glucagon-like peptide-1 in these cells. These results indicate that HGF reduces the acinar cell-like property of AR42J cells and converts them into insulin-producing cells. The effect of HGF was markedly enhanced by activin A.

Journal ArticleDOI
TL;DR: Two Met-mediated biological responses, motility and growth, can be dissociated on the basis of their differential requirement for a direct link with Ras.

Journal ArticleDOI
TL;DR: It is concluded that dHGF prevents and improves the DMN‐induced hepatic fibrosis in rats by reducing mRNA levels of procollagens and TGF‐β1, by inhibiting an activation of hepatic lipocytes, and by stimulating liver regeneration.

Journal ArticleDOI
TL;DR: It is shown that ectopic application of exogenous SF/HGF induces emigration of Pax-3-positive myogenic cells into the lateral plate mesoderm and plays a necessary role in the initiation of myoblast migration.

Journal ArticleDOI
TL;DR: Results indicate that HGF is a potent mitogen for airway epithelial cells and alveolar epithelial Cells in vivo as well as in vitro, and may act as pulmotrophic factor responsible forAirway andAlveolar regeneration during lung regeneration after acute lung injury.
Abstract: Hepatocyte growth factor (HGF) has mitogenic, morphogenic, and motogenic activities on epithelial cells and plays important roles in regeneration of the liver and the kidney. We previously found that the expression of HGF gene is rapidly induced in the lung after acute lung injury in experimental animals and that HGF levels are elevated in blood of patients with lung diseases. To search for a possible pulmotrophic function of HGF in lung regeneration, we examined the mitogenic activity of HGF on tracheal epithelial cells in vitro and evaluated the efficacy of HGF-administration on lung regeneration after acute lung injury in mice. HGF markedly stimulated proliferation and DNA synthesis of rat tracheal epithelial cells in primary culture in a dose-dependent manner. The intravenous injection of human recombinant HGF (10 micrograms.mouse-1.day-1) into mice with acute lung injury induced by the intratracheal infusion of 10 mM HCI stimulated DNA synthesis of airway epithelial cells to levels threefold higher than those in mice with no HGF-injections, but it did not stimulate DNA synthesis of alveolar epithelial cells. However, HGF injection at higher dose (100 micrograms.mouse-1.day-1) stimulated DNA synthesis of alveolar epithelial cells in vivo. These results indicate that HGF is a potent mitogen for airway epithelial cells and alveolar epithelial cells in vivo as well as in vitro. HGF may act as pulmotrophic factor responsible for airway and alveolar regeneration during lung regeneration after acute lung injury.

Journal Article
TL;DR: Comparison of transgenic and control liver morphology revealed dramatic heterogeneity in the size and appearance of hepatocytes as a distinctive feature of HGF/SF overexpression, and the mechanistic basis of hepatocyte proliferation was elucidated as the chronic activation of the c-met proto-oncogene product.
Abstract: To investigate the in vivo role of hepatocyte growth factor/scatter factor (HGF/SF) in liver function, we generated transgenic mice using a mouse HGF/SF cDNA under the control of the mouse metallothionein gene promoter and 5'/3' flanking sequences. In adult HGF/SF transgenic mice, liver weight as a percentage of total body weight was at least twice that of wild-type mice. Comparison of transgenic and control liver morphology revealed dramatic heterogeneity in the size and appearance of hepatocytes as a distinctive feature of HGF/SF overexpression. Transgenic livers exhibited a significant increase in the number of small hepatocytes with a 2N DNA content, accounting for the observed increase in liver mass. The DNA labeling index of hepatocytes increased 11-fold at 4 weeks of age, when liver enlargement first became apparent, and was still elevated about 5-fold in adult HGF/SF transgenic mice. Moreover, hepatocytes isolated by perfusion of transgenic livers doubled every 2 days in culture, whereas little or no growth was observed with isolated control hepatocytes. The mechanistic basis of hepatocyte proliferation was elucidated as the chronic activation of the c-met proto-oncogene product. Met and substrates such as phosphatidylinositol 3-kinase, Src homology and collagen-like, pp60c-src, focal adhesion kinase p125FAK, and paxillin were associated with tyrosine-phosphorylated complexes in a hepatocyte cell line established from the transgenic liver. This proliferative stimulus triggered the formation of hepatocellular adenomas and/or carcinomas in most transgenic mice > or = 1.5 years of age. Finally, the rate of transgenic mouse liver regeneration was increased 3-fold over control livers following partial hepatectomy.

Journal ArticleDOI
TL;DR: Examination of the expression of the met/HGF receptor in normal human kidney and tissues of the genito‐urinary tract, and in 50 kidney neoplasms of different histotypes, suggests that expression of this receptor may be involved in the onset and progression of renal cell carcinomas.
Abstract: The c-met oncogene encodes the receptor for hepatocyte growth factor/scatter factor (HGF/SF), a multifunctional cytokine able to mediate morphogenesis as well as mitogenesis, motogenesis and invasiveness of epithelial cells. HGF/SF has been implicated in branching tubulogenesis of the developing kidney and in regeneration after renal injury and nephrectomy. We have examined the expression of the met/HGF receptor in normal human kidney and tissues of the genito-urinary tract, and in 50 kidney neoplasms of different histotypes, using monoclonal antibodies (MAbs) against the met/HGF receptor and immunohistochemistry. In normal kidneys, weak staining restricted to the distal tubules was observed. Transitional cell carcinomas were consistently negative, whereas increased expression at various levels was found in 87% of renal cell carcinomas with different cytological features and histological patterns. Western blot analysis of samples showed that the met/HGF receptor found in the malignant cells exhibits features of the normal receptor. The met/HGF receptor is also overexpressed in a renal cell carcinoma cell line, whose motility is triggered by HGF/SF. Our data suggest that expression of the met/HGF receptor may be involved in the onset and progression of renal cell carcinomas.

Journal ArticleDOI
TL;DR: It is proposed that the normal agonist activity of NK1 and NK2 in mink lung cells is due to an activating interaction with an endogenous glycosaminoglycans, and that a large portion of the NK1 binding to mink Lung cells could be blocked by heparin.
Abstract: Hepatocyte growth factor (HGF) is a potent epithelial mitogen whose actions are mediated through its receptor, the proto-oncogene c-Met. Two truncated variants of HGF known as NK1 and NK2 have been reported to be competitive inhibitors of HGF binding to c-Met, and to function as HGF antagonists (Lokker, N.A., and P.J. Godowski. 1993. J. Biol. Chem. 268: 17145-17150; Chan, A.M., J.S. Rubin, D.P. Bottaro, D.W. Hirschfield, M. Chedid, and S.A. Aaronson. 1991. Science (Wash. DC). 254:1382-1387). We show here, however, that NK1 acts as a partial agonist in mink lung cells. Interestingly, NK1, which is an HGF antagonist in hepatocytes in normal conditions, was converted to a partial agonist by adding heparin to the culture medium. The interaction of NK1 and heparin was further studied in BaF3 cells, which express little or no cell surface heparan sulfate proteoglycans. In BaF3 cells transfected with a plasmid encoding human c-Met, heparin and NK1 synergized to stimulate DNA synthesis and cell proliferation. There was no effect of heparin on the IL-3 sensitivity of BaF3-hMet cells, and no effect of NK1 plus heparin in control BaF3 cells, indicating that the response was specific and mediated through c-Met. The naturally occurring HGF splice variant NK2 also stimulated DNA synthesis in mink lung cells and exerted a heparin-dependent effect on BaF3-hMet cells, but not on BaF3-neo cells. The activating effect of heparin was mimicked by a variety of sulfated glycosaminoglycans. Mechanistic studies revealed that heparin increased the binding of NK1 to BaF3-hMet cells, stabilized NK1, and induced dimerization of NK1. Based on these studies, we propose that the normal agonist activity of NK1 and NK2 in mink lung cells is due to an activating interaction with an endogenous glycosaminoglycan. Consistent with that model, a large portion of the NK1 binding to mink lung cells could be blocked by heparin. Moreover, a preparation of glycosaminoglycans from the surface of mink lung cells induced dimerization of NK1. These data show that the activity of NK1 and NK2 can be modulated by heparin and other related glycosaminoglycans to induce proliferation in cells expressing c-Met.

Journal ArticleDOI
TL;DR: It is proposed that in homozygous Splotch embryos the failure of muscle precursors to migrate into and populate the limb bud results from a loss of met expression in the cells at the ventrolateral edge of the somitic dermomyotome.
Abstract: Hepatocyte growth factor/scatter factor (HGF/SF) stimulates proliferation, dissociation, migration and morphogenesis of cells in culture. To investigate a possible role for HGF/SF and its receptor, the Met tyrosine kinase, in embryonic development, we have analyzed their expression in mouse embryos from day 7.5 of gestation by whole-mount in situ hybridization. Met expression is first detected in the ventral portion of somites at day 9.25 of gestation (22 somite embryo) at the level of fore limb buds. As somites mature, met expression is detected in caudal somites, and is confined to the lateral and media] tips of the dermomyotome and dermomyotome/myotome respectively. In contrast, HGF/SF is expressed exclusively in the mesodermal core of the limb bud. As the dermomyotome elongates ventrolaterally, the met-expressing cells at the lateral tip appear to detach from the somite, invade the limb bud and localize at the dorsal and ventral limb sides in close proximity to HGF/SF-expressing cells. At later stages, both met- and HGF/SF-expressing cells appear to migrate distally and localize to the digit forming area of the developing hand plate. Met expression in the lateral dermomyotome and limb bud coincides with expression of Pax-3, a marker for migrating muscle precursor cells in the somite and limb. Splotch-2H and Splotch-delayed mice, which harbor mutations in Pax-3, show major disruptions in early limb muscle development. Significantly, no met-expressing cells were observed in the limbs of homozygous Splotch-2H and Splotch-delayed animals, whereas HGF/SF expression was not affected. The restricted expression of met to a sub-population of Pax-3-expressing cells in the lateral tip of the dermomyotome, demonstrates that met represents a unique molecular marker for this migratory cell population. From these observations, together with the biological activities of HGF/SF, we propose that in homozygous Splotch embryos the failure of muscle precursors to migrate into and populate the limb bud results from a loss of met expression in the cells at the ventrolateral edge of the somitic dermomyotome.