201
|
Wu JH, Billings BJ, Balkovetz DF. Hepatocyte growth factor alters renal epithelial cell susceptibility to uropathogenic Escherichia coli. J Am Soc Nephrol 2001; 12:2543-2553. [PMID: 11729222 DOI: 10.1681/asn.v12122543] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The urinary tract is frequently the source of Escherichia coli bacteremia. Bacteria from the urinary tract must cross an epithelial layer to enter the bloodstream. Hepatocyte growth factor (HGF) alters the polarity of Madin-Darby canine kidney (MDCK) epithelial cells. The role of cell polarity in determining renal epithelial resistance to Escherichia coli invasion is not well known. A model of polarized and HGF-treated MDCK epithelial cells grown on filters was used to study the role of epithelial cell polarity during the interaction of nonvirulent (XL1-Blue) and uropathogenic (J96) strains of Escherichia coli with renal epithelium. Basolateral exposure of MDCK cells to J96, but not XL1-Blue, resulted in loss of transepithelial resistance (TER), which was due to epithelial cytotoxicity and not degradation of epithelial junctional proteins by bacterial proteases. Apical exposure to both J96 and XL1-Blue did not alter TER. Pretreatment of polarized MDCK cell monolayers with HGF renders the cells sensitive to loss of TER and cytotoxicity by apical exposure to J96. Analysis by confocal microscopy demonstrated that HGF treatment of MDCK cell monolayers also greatly enhances adherence of J96 to the apical surface of the cell monolayer. These data demonstrate that the basolateral surface of polarized epithelia is more susceptible to J96 cytotoxicity. The data also support the hypothesis that processes that alter epithelial cell polarity increase sensitivity of epithelia to bacterial injury and adherence from the apical compartment.
Collapse
Affiliation(s)
- John H Wu
- *Birmingham Veterans Affairs Medical Center, Birmingham, Alabama; and Departments of Medicine, Cell Biology, and Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Barry J Billings
- *Birmingham Veterans Affairs Medical Center, Birmingham, Alabama; and Departments of Medicine, Cell Biology, and Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel F Balkovetz
- *Birmingham Veterans Affairs Medical Center, Birmingham, Alabama; and Departments of Medicine, Cell Biology, and Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
202
|
Deng J, Kohda Y, Chiao H, Wang Y, Hu X, Hewitt SM, Miyaji T, McLeroy P, Nibhanupudy B, Li S, Star RA. Interleukin-10 inhibits ischemic and cisplatin-induced acute renal injury. Kidney Int 2001; 60:2118-28. [PMID: 11737586 DOI: 10.1046/j.1523-1755.2001.00043.x] [Citation(s) in RCA: 313] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Acute renal failure (ARF) is caused by ischemic and nephrotoxic insults acting alone or in combination. Anti-inflammatory agents have been shown to decrease renal ischemia-reperfusion and cisplatin-induced injury and leukocyte infiltration. Interleukin-10 (IL-10) is a potent anti-inflammatory cytokine that inhibits inflammatory and cytotoxic pathways implicated in acute renal injury. Therefore, we sought to determine if IL-10 inhibits acute renal injury. METHODS The effects of IL-10 were studied in mice following cisplatin administration and bilateral renal ischemia-reperfusion, in a rat model of renal transplantation, and in cultured mouse cortical tubule cells. RESULTS IL-10 significantly decreased renal injury following cisplatin administration and following renal ischemia/reperfusion. Delay of IL-10 treatment for one hour after cisplatin also significantly inhibited renal damage. IL-10 and alpha-melanocyte stimulating hormone (alpha-MSH) increased recovery following transplantation of a kidney subjected to warm ischemia. To explore the mechanism of action of IL-10, its effects were measured on mediators of leukocyte trafficking and inducible nitric oxide synthase (NOS-II). IL-10 inhibited cisplatin and ischemia-induced increases in mRNA for tumor necrosis factor-alpha (TNF-alpha), intercellular adhesion molecule-1 (ICAM-1), and NOS-II. IL-10 also inhibited staining for markers of apoptosis and cell cycle activity following cisplatin administration, and nitric oxide production in cultured mouse cortical tubules. CONCLUSIONS IL-10 protects against renal ischemic and cisplatin-induced injury. IL-10 may act, in part, by inhibiting the maladaptive activation of genes that cause leukocyte activation and adhesion, and induction of iNOS.
Collapse
Affiliation(s)
- J Deng
- Renal Diagnostics and Therapeutics Unit, NIDDK, and Laboratory of Pathology, NCI, National Institutes of Health, Bethesda, Maryland 20892-1268, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Ernst F, Hetzel S, Stracke S, Czock D, Vargas G, Lutz MP, Keller F, Jehle PM. Renal proximal tubular cell growth and differentiation are differentially modulated by renotropic growth factors and tyrosine kinase inhibitors. Eur J Clin Invest 2001; 31:1029-39. [PMID: 11903488 DOI: 10.1046/j.1365-2362.2001.00925.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The renotropic growth factors (GFs), hepatocyte GF (HGF), epidermal GF (EGF), and insulin-like GF-I (IGF-I) accelerate renal regeneration in animal models after toxic or ischemic injury. These GFs initiate their biological effects on renal tubular cells by interaction with specific transmembrane receptor tyrosine kinases. MATERIALS AND METHODS In the proximal tubular cell line PT-1, the biological effects of HGF, EGF, and IGF-I and the growth-inhibitory effects of different tyrosine kinase inhibitors (TKIs) were investigated. Receptor binding and tyrosine kinase phosphorylation were determined by ligand binding studies and Western blot analysis. RESULTS HGF, EGF, and IGF-I bound with nanomolar affinity to their specific cell membrane receptor tyrosine kinases. In contrast to EGF or IGF-I, HGF induced a variety of cell morphological changes, including cell scattering, formation of tubular structures, and expression of long microvilli on the apical cell membrane. HGF was a 10-fold more potent and more effective growth promoter than EGF or IGF-I. Among the TKIs tested, the mitogenic effect of HGF could be more specifically inhibited by emodin and tyrphostin, that of EGF by methyl-2,5-dihydroxycinnamate, lavendustin A, and genistein, and that of IGF-I by geldanamycin. CONCLUSIONS In contrast to EGF and IGF-I, HGF stimulated both growth and differentiation of renal proximal tubular cells, demonstrating the amazing biological potency of this renotropic growth factor. Selective TKIs may be a promising approach to modulate diseases with abnormalities in protein kinase signalling pathways such as renal cell carcinoma.
Collapse
Affiliation(s)
- F Ernst
- Division of Nephrology, Department of Internal Medicine II, University of Ulm, Robert-Koch-Strasse 8, D-89081 Ulm, Germany
| | | | | | | | | | | | | | | |
Collapse
|
204
|
Haug C, Schmid-Kotsas A, Linder T, Bachem MG, Gruenert A, Rozdzinski E. Influence of hepatocyte growth factor, epidermal growth factor, and mycophenolic acid on endothelin-1 synthesis in human endothelial cells. Nephrol Dial Transplant 2001; 16:2310-6. [PMID: 11733621 DOI: 10.1093/ndt/16.12.2310] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Endothelin-1 (ET-1) is a potent vasoconstrictive peptide which plays an important pathophysiological role in ischaemic renal failure and drug-induced renal injury such as cyclosporin A (CsA)- and tacrolimus-associated nephrotoxicity. In contrast, hepatocyte growth factor (HGF) and epidermal growth factor (EGF) seem to accelerate renal regeneration after ischaemic and drug-induced renal injury. This study aimed to investigate the influence of HGF and EGF on ET-1 synthesis in cultured human umbilical vein endothelial cells (HUVEC) and renal artery endothelial cells (RAEC). In addition, we have investigated whether mycophenolic acid (MPA), a new immunosuppressive drug, which in contrast to CsA and tacrolimus lacks nephrotoxic side effects, modulates ET-1 synthesis in endothelial cells. METHODS ET-1 release was measured with a specific enzyme-linked immunosorbent assay. ET-1 mRNA expression was investigated by reverse transcription polymerase chain reaction. RESULTS HGF and EGF (0.001-10 nM) exerted a significant concentration-dependent inhibitory effect on ET-1 release by HUVEC and RAEC (minimum 56.1+/-4.3% of control, n=6, mean+/-SE). The suppressive effect of HGF and EGF on ET-1 synthesis was dose-dependently antagonized by the tyrosine kinase inhibitors tyrphostin AG1478, lavendustin A and methyl 2,5-dihydroxycinnamate. Incubation of HUVEC and RAEC with MPA (2.5, 10, 25, and 50 microg/ml) for 3-5 h induced a significant reduction of ET-1 mRNA expression. After 48 h incubation with MPA (1-50 microg/ml) a significant decrease of ET-1 release and DNA content per culture well was observed, whereas ET-1 release referred to the DNA content in the corresponding culture well did not differ significantly from controls. CONCLUSIONS The present findings demonstrate that HGF and EGF reduce ET-1 synthesis in endothelial cells via their receptor tyrosine kinase activity and suggest that the renoprotective effects of HGF and EGF might be linked to their inhibitory action on ET-1 synthesis. This study also provides evidence that, in contrast to CsA and tacrolimus, MPA does not stimulate ET-1 synthesis. This might explain the clinical observation that renal function often improves when CsA or tacrolimus is replaced by mycophenolate mofetil.
Collapse
Affiliation(s)
- C Haug
- Institute of Clinical Chemistry, University Hospital, Ulm, Germany.
| | | | | | | | | | | |
Collapse
|
205
|
Nakamura T, Kanda S, Yamamoto K, Kohno T, Maeda K, Matsuyama T, Kanetake H. Increase in hepatocyte growth factor receptor tyrosine kinase activity in renal carcinoma cells is associated with increased motility partly through phosphoinositide 3-kinase activation. Oncogene 2001; 20:7610-23. [PMID: 11753639 DOI: 10.1038/sj.onc.1204975] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2001] [Revised: 08/28/2001] [Accepted: 09/13/2001] [Indexed: 11/08/2022]
Abstract
Dysregulated cell motility is one of the major characteristics of invasion and metastatic potentials of malignant tumor cells. Here, we examined the hepatocyte growth factor (HGF)-induced cell motility of two human renal carcinoma cell lines, ACHN and VMRC-RCW. Scattering and migration was induced in ACHN in an HGF-dependent manner, whereas they were maintained in VMRC-RCW even in the absence of HGF. In VMRC-RCW, HGF receptor (HGFR) tyrosine kinase was constitutively active, and sequence analysis showed N375S, A1209G and V1290L mutations. However, transfection experiments using porcine aortic endothelial (PAE) cells demonstrated that no single mutation or combination of two or three mutations caused HGF-independent constitutive activation. Conversely, the expressed amount of receptor protein had a pivotal role in the basal kinase activity. With respect to downstream signaling molecules of HGFR in ACHN or VMRC-RCW, the Ras-MAPK pathway was downregulated, whereas phosphoinositide 3-kinase (PI3-kinase) was not further activated by HGF-treatment in VMRC-RCW cells. The PI3-kinase inhibitors, wortmannin and LY294002 strongly inhibited spontaneous migration of VMRC-RCW. One transfected PAE cell line with massive overexpression of HGFR demonstrated scattered morphology and increased PI3-kinase activity in association with increased motility, which was partially inhibited by LY294002. Taken together, our results indicate that the overexpression of HGFR causes increase in cellular motility and PI3-kinase shows the important contribution on the increased motility of renal carcinoma cells.
Collapse
Affiliation(s)
- T Nakamura
- Department of Urology, Nagasaki University School of Medicine, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
206
|
Yang J, Dai C, Liu Y. Systemic administration of naked plasmid encoding hepatocyte growth factor ameliorates chronic renal fibrosis in mice. Gene Ther 2001; 8:1470-1479. [PMID: 11593360 DOI: 10.1038/sj.gt.3301545] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2001] [Accepted: 07/05/2001] [Indexed: 11/08/2022]
Abstract
The progression of chronic renal diseases is considered as an irreversible process that eventually leads to end-stage renal failure characterized by extensive tissue fibrosis. At present, chronic renal fibrosis is incurable and the incidence of affected patients is on the rise worldwide. In this study, we demonstrate that delivery of hepatocyte growth factor (HGF) gene via systemic administration of naked plasmid vector markedly ameliorated renal fibrosis in an animal model of chronic renal disease induced by unilateral ureteral obstruction. A high level of exogenous HGF protein was detected in the obstructed kidneys following intravenous injection of naked plasmid encoding human HGF. Delivery of human HGF gene induced a sustained activation of extracellular signal-regulated kinases-1 and -2 in the obstructed kidneys. Exogenous HGF expression dramatically inhibited alpha-smooth muscle actin expression, attenuated renal interstitial accumulation and deposition of collagen I and fibronectin. In addition, exogenous HGF suppressed renal expression of pro-fibrogenic cytokine TGF-beta1 and its type I receptor in vivo. These results suggest that systemic administration of naked plasmid vector introduces a high level of exogenous HGF to the diseased kidneys, and that HGF gene transfer may provide a novel therapeutic strategy for amelioration of chronic renal fibrosis in vivo.
Collapse
Affiliation(s)
- J Yang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
207
|
Fernández M, Medina A, Santos F, Carbajo E, Rodríguez J, Álvarez J, Cobo A. Exacerbated inflammatory response induced by insulin-like growth factor I treatment in rats with ischemic acute renal failure. J Am Soc Nephrol 2001; 12:1900-1907. [PMID: 11518783 DOI: 10.1681/asn.v1291900] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
In agreement with recent studies showing a deleterious effect of growth hormone treatment in critically ill patients, preliminary data showed that insulin-like growth factor I (IGF-I) administration increased the mortality rate of rats with ischemic acute renal failure (ARF). The present study was designed to investigate the mechanism responsible for this unexpected effect. Male rats with ischemic ARF were given subcutaneous IGF-I, 50 microg/100 g at 0, 8, and 16 h after reperfusion (ARF+IGF-I, n = 5) or were untreated (ARF, n = 5). A group of 5 sham-operated rats were used as controls. Rats were killed 48 h after declamping, and the following studies were performed: in serum, creatinine and urea nitrogen; and in kidneys, histologic damage score, cellular proliferation by bromodeoxyuridine labeling, apoptosis by morphologic criteria, macrophage infiltration by immunohistochemistry using a specific antibody against ED-1, neutrophil infiltration by naphthol AS-D chloroacetate esterase staining, and levels of IGF-I and IGF-I receptor mRNA by RNase protection assay. ARF and ARF+IGF-I groups had a severe and similar degree of renal failure. Kidney damage was histologically more evident in ARF+IGF-I (1.9 +/- 0.1) than in ARF (1.3 +/- 0.2) rats, and the number of neutrophils/mm(2) of tissue was significantly greater in ARF+IGF-I than in ARF rats at the corticomedullary junction (52.3 +/- 5.2 versus 37.2 +/- 4.1) as well as at the renal medulla (172.5 +/- 30.0 versus 42.1 +/- 9.6). No other differences between the groups were found. It is concluded that IGF-I treatment enhanced the inflammatory response in rats with ischemic ARF. Cell toxicity derived from increased neutrophil accumulation might play a key role in the greater mortality risk of critically ill patients that are treated with growth hormone.
Collapse
Affiliation(s)
- Marta Fernández
- Department of Pediatrics, Hospital Central de Asturias and School of Medicine, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Asturias, Spain
| | - Alberto Medina
- Department of Pediatrics, Hospital Central de Asturias and School of Medicine, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Asturias, Spain
| | - Fernando Santos
- Department of Pediatrics, Hospital Central de Asturias and School of Medicine, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Asturias, Spain
| | - Eduardo Carbajo
- Department of Anatomy, Hospital Central de Asturias and School of Medicine, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Asturias, Spain
| | - Julián Rodríguez
- Department of Pediatrics, Hospital Central de Asturias and School of Medicine, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Asturias, Spain
| | - Jesús Álvarez
- Department of Pediatrics, Hospital Central de Asturias and School of Medicine, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Asturias, Spain
| | - Angeles Cobo
- Department of Pediatrics, Hospital Central de Asturias and School of Medicine, Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Asturias, Spain
| |
Collapse
|
208
|
Azuma H, Takahara S, Matsumoto K, Ichimaru N, Wang JD, Moriyama T, Waaga AM, Kitamura M, Otsuki Y, Okuyama A, Katsuoka Y, Chandraker A, Sayegh MH, Nakamura T. Hepatocyte growth factor prevents the development of chronic allograft nephropathy in rats. J Am Soc Nephrol 2001; 12:1280-1292. [PMID: 11373353 DOI: 10.1681/asn.v1261280] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Long-term renal isografts in humans and laboratory animals exhibit features similar to those of chronic allograft nephropathy (CAN), indicating that antigen-independent factors, such as acute renal ischemia, are likely to be involved in the development of CAN. Hepatocyte growth factor (HGF) has been demonstrated to play a renotropic role in renal regeneration and protection from acute ischemic injury. This study was thus conducted to investigate the effect of HGF on the development of CAN, using an established rat model. HGF was administered daily (100 microg/d, intravenously) for 4 wk after engraftment. Control animals received saline solution. Allografts from control animals exhibited early evidence of severe structural collapse and necrotic cell death in the proximal tubules and outer medulla, with mononuclear cell infiltration, within 1 wk after engraftment. This was followed by sequential upregulation of adhesion molecules and cytokines, accompanied by dense macrophage infiltration. Fibrogenic events, as indicated by marked increases in transforming growth factor-beta1 expression and the accumulation of smooth muscle alpha-actin, occurred during the same period. Control animals ultimately developed features typical of CAN, with functional deterioration and severe histologic changes; a survival rate of 50.6% by 32 wk was observed. In contrast, remarkably little early injury and no late fibrogenic events were observed for the HGF-treated group. All treated animals survived, with well preserved graft function, during the 32-wk follow-up period. These results indicate that renal protection and recovery from early allograft injury with HGF treatment greatly contribute to a reduction of susceptibility to the subsequent development of CAN in a rat model. The potential application of HGF in the prevention of CAN warrants further attention.
Collapse
Affiliation(s)
- Haruhito Azuma
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shiro Takahara
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kunio Matsumoto
- Division of Biochemistry, Biomedical Research Center, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naotsugu Ichimaru
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jing Ding Wang
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshiki Moriyama
- Department of Internal Medicine and Therapeutics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ana-Maria Waaga
- Laboratory of Immunogenetics and Transplantation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Masaya Kitamura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshinori Otsuki
- Department of Anatomy and Biology, Osaka Medical College, Osaka, Japan
| | - Akihiko Okuyama
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoji Katsuoka
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Anil Chandraker
- Laboratory of Immunogenetics and Transplantation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mohamed H Sayegh
- Laboratory of Immunogenetics and Transplantation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Toshikazu Nakamura
- Division of Biochemistry, Biomedical Research Center, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
209
|
Warn R, Harvey P, Warn A, Foley-Comer A, Heldin P, Versnel M, Arakaki N, Daikuhara Y, Laurent GJ, Herrick SE, Mutsaers SE. HGF/SF induces mesothelial cell migration and proliferation by autocrine and paracrine pathways. Exp Cell Res 2001; 267:258-66. [PMID: 11426944 DOI: 10.1006/excr.2001.5240] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mesothelial repair differs from that of other epithelial-like surfaces as healing does not occur solely by centripetal in-growth of cells as a sheet from the wound margins. Mesothelial cells lose their cell-cell junctions, divide, and adopt a fibroblast-like morphology while scattering across and covering the wound surface. These features are consistent with a cellular response to hepatocyte growth factor/scatter factor (HGF/SF). In this study, we examined the ability of mesothelial cells to secrete HGF/SF and investigated its possible role as an autocrine regulator of mesothelial cell motility and proliferation. We found that human primary mesothelial cells expressed HGF/SF mRNA and secreted active HGF/SF into conditioned medium as determined by ELISA and in a scattering bioassay. These cells also expressed the HGF/SF receptor, Met, as shown by RT-PCR and by Western blot analysis and immunofluorescence. Incubation of mesothelial cells with neutralizing antibodies to HGF/SF decreased cell migration to 25% of controls, whereas addition of HGF/SF disrupted cell-cell junctions and induced scattering and enhanced mesothelial cell migration. Furthermore, HGF/SF showed a small but significant mitogenic effect on all mesothelial cell lines examined. In conclusion, HGF/SF is produced by mesothelial cells and induces both motility and proliferation of these cells. These data are consistent with HGF/SF playing an autocrine role in mesothelial healing.
Collapse
Affiliation(s)
- R Warn
- School of Biology, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Okamoto T, Takatsuka H, Fujimori Y, Wada H, Iwasaki T, Kakishita E. Increased hepatocyte growth factor in serum in acute graft-versus-host disease. Bone Marrow Transplant 2001; 28:197-200. [PMID: 11509938 DOI: 10.1038/sj.bmt.1703095] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2001] [Accepted: 04/26/2001] [Indexed: 12/22/2022]
Abstract
Hepatocyte growth factor (HGF) was reported to be effective in preventing acute graft-versus-host disease (GVHD) in a murine model. We examined serum HGF concentrations in 38 patients receiving allogeneic bone marrow transplants, and investigated the relationship of serum HGF concentrations to severity of acute GVHD. More HGF was present in sera from patients with than without acute GVHD. Serum HGF correlated significantly with grade of acute GVHD. Furthermore, serum HGF correlated with serum concentrations of C-reactive protein, gamma-glutamyltranspeptidase (GTP), and aspartate aminotransferase (AST). Serum concentrations of HGF in transplanted patients without GVHD were consistently low, while those in patients with acute GVHD increased with exacerbation. We conclude that HGF was produced during induction of the GVH reaction, and probably increased as a physiological response.
Collapse
Affiliation(s)
- T Okamoto
- Second Department of Internal Medicine, Hyogo College of Medicine, Hyogo, Japan
| | | | | | | | | | | |
Collapse
|
211
|
Liu ZX, Nickel CH, Cantley LG. HGF promotes adhesion of ATP-depleted renal tubular epithelial cells in a MAPK-dependent manner. Am J Physiol Renal Physiol 2001; 281:F62-70. [PMID: 11399647 DOI: 10.1152/ajprenal.2001.281.1.f62] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hepatocyte growth factor (HGF) has been shown to enhance recovery from renal tubular ischemia. We investigated the possibility that HGF improves recovery by preventing ischemia-induced loss of cell adhesion. Murine inner medullary collecting duct-3 (mIMCD-3) cells subjected to 90% ATP depletion demonstrated a 55% decrease in adhesion, an effect that was completely reversed by the addition of HGF. Assays examining release of adherent cells revealed similar results with 30 min of ATP depletion causing loss of adhesion of 25% of mIMCD-3 cells and HGF completely reversing this effect. In contrast, HGF was unable to reverse the loss of adhesion of cells exposed to 99% ATP depletion. Examination of the mitogen-activated protein kinase (MAPK) signaling pathway revealed that HGF could induce extracellular signal-regulated kinase (ERK) phosphorylation in control and 90% ATP-depleted cells but not in 99% ATP-depleted cells. Inhibition of ERK activation with U0126 completely blocked the HGF-dependent reversal of ATP-depleted cell adhesion. Thus ATP-depleted cells demonstrate a marked decrease in cell adhesion that is reversible by the addition of HGF. This effect of HGF requires activation of the MAPK pathway.
Collapse
Affiliation(s)
- Z X Liu
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | | | | |
Collapse
|
212
|
Kuroiwa T, Kakishita E, Hamano T, Kataoka Y, Seto Y, Iwata N, Kaneda Y, Matsumoto K, Nakamura T, Ueki T, Fujimoto J, Iwasaki T. Hepatocyte growth factor ameliorates acute graft-versus-host disease and promotes hematopoietic function. J Clin Invest 2001; 107:1365-73. [PMID: 11390418 PMCID: PMC209319 DOI: 10.1172/jci11808] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Acute graft-versus-host disease (GVHD) is a major complication of bone marrow transplantation (BMT) and is characterized by hematopoietic dysfunction, immunosuppression, and tissue injury in the skin, liver, and intestinal mucosa. Hepatocyte growth factor (HGF), originally identified and cloned as a potent mitogen for hepatocytes, induces mitogenic and antiapoptotic activity in various epithelial cells and promotes hematopoiesis. Working in a murine model of acute GVHD, we performed repeated transfection of the human HGF cDNA into skeletal muscle and showed that this treatment inhibited apoptosis of intestinal epithelial cells and donor T-cell infiltration into the liver, thereby ameliorating the enteropathy and liver injury caused by acute GVHD. HGF also markedly suppressed IFN-gamma and TNF-alpha expression in the intestine and liver and decreased the serum IL-12. Furthermore, extramedullary hematopoiesis by donor cells was increased, and the survival rate was improved. These results suggest that HGF may be useful for controlling acute GVHD after allogeneic BMT.
Collapse
Affiliation(s)
- T Kuroiwa
- Second Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Matsumoto K, Nakamura T. Hepatocyte growth factor: renotropic role and potential therapeutics for renal diseases. Kidney Int 2001; 59:2023-38. [PMID: 11380804 DOI: 10.1046/j.1523-1755.2001.00717.x] [Citation(s) in RCA: 254] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hepatocyte growth factor (HGF), a ligand for the c-Met receptor tyrosine kinase, has mitogenic, motogenic, anti-apoptotic, and morphogenic (for example, induction of branching tubulogenesis) activities for renal tubular cells, while it has angiogenic and angioprotective actions for endothelial cells. Stromal cells such as mesangial cells, endothelial cells, and macrophages are sources of renal HGF; thus, HGF mediates epithelial-stromal and endothelial-mesangial interactions in the kidney. In response to acute renal injury, the expression of HGF increases in the injured kidney and in distant intact organs such as the lung and spleen. Locally and systemically increased HGF supports renal regeneration, possibly not only by enhancing cell growth but also by promoting morphogenesis of renal tissue. During progression of chronic renal failure/renal fibrosis, the expression of HGF decreases in a manner reciprocal to the increase in expression of transforming growth factor-beta (TGF-beta), a key player in tissue fibrosis. A decrease in endogenous HGF, as well as increase in TGF-beta, augments susceptibility to the onset of chronic renal failure/renal fibrosis. On the other hand, supplements of exogenous HGF have preventive and therapeutic effects in cases of acute and chronic renal failure/renal fibrosis in laboratory animals. HGF prevents epithelial cell death and enhances regeneration and remodeling of renal tissue with injury or fibrosis. A renotropic system underlies the vital potential of the kidney to regenerate, while an impaired renotropic system may confer susceptibility to the onset of renal diseases. Thus, HGF supplementation may be one therapeutic strategy to treat subjects with renal diseases, as it enhances the intrinsic ability of the kidney to regenerate.
Collapse
Affiliation(s)
- K Matsumoto
- Division of Biochemistry, Biomedical Research Center, Osaka University Graduate School of Medicine, Osaka, Japan.
| | | |
Collapse
|
214
|
Sugimura K, Goto T, Tsuchida K, Takemoto Y, Kim T, Kishimoto T. Production and activation of hepatocyte growth factor in acute renal failure. Ren Fail 2001; 23:597-603. [PMID: 11499573 DOI: 10.1081/jdi-100104741] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hepatocyte growth factor (HGF) facilitates the regeneration of injured kidney in acute renal failure (ARF). HGF is produced as a single-chain precursor by cells of mesenchymal origin and is converted to a biologically active, heterodimeric molecule by proteolytic processing. We studied HGF mRNA and protein levels in systemic organs of glycerol-induced ARF rats, a model of crush syndrome. HGF protein concentration of tissue homogenate was measured by ELISA. Both mRNA and protein levels were increased in liver and spleen at 24 hours after the glycerol injection whereas HGF protein level was decreased in the injured kidney. Expression of HGF receptor/c-met mRNA was elevated only in the kidney. These results suggest that HGF supplied in an endocrine manner may play an important role in the regenerating process following ARF. Next, we measured serum HGF concentration by ELISA in 8 ARF patients caused by crush syndrome and the molecular size of serum HGF was determined by immunoblotting. Although serum HGF levels elevated in all patients, the HGF levels did not associate with their prognoses. While a single-chain molecule was predominantly observed in sera from chronic renal failure patients and healthy subjects, the majority of serum HGF was a heterodimeric form in 7 ARF patients. In one patient who developed disseminated intravascular coagulation syndrome and had a poor prognosis, a single-chain molecule was predominant although the serum HGF concentration was equivalent. These data suggest that the activity of proteolytic processing may be also an important factor for the expression of the biological function of HGF.
Collapse
Affiliation(s)
- K Sugimura
- Department of Urology, Osaka City University Medical School, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
215
|
Abstract
Scatter factors are unequivocal signals governing a genetic program that includes cell detachment, repulsion, protection from apoptosis, invasiveness of extracellular matrices and proliferation. This pleiomorphic response is defined as 'invasive growth'. Under physiological conditions, it leads to morphogenic cell movements through the matrix, and--primarily--to ordered building of epithelial tubules. Dysfunctions in invasive growth cause enhanced proliferation, uncontrolled migration into surrounding tissues, and failure to differentiate, events that foster tumour growth and invasiveness. Scatter factors act through tyrosine kinase receptors that belong to the Met oncogene family. Here we discuss how alterations of these receptors or of their signal transduction pathways are responsible for cancer onset and progression towards metastasis.
Collapse
Affiliation(s)
- P M Comoglio
- Division of Molecular Oncology, IRCC, Institute for Cancer Research, Str. Provinciale 142, km. 3.95, Candiolo (TO), 10060, Italy.
| | | |
Collapse
|
216
|
Yang J, Chen S, Huang L, Michalopoulos GK, Liu Y. Sustained expression of naked plasmid DNA encoding hepatocyte growth factor in mice promotes liver and overall body growth. Hepatology 2001; 33:848-859. [PMID: 11283849 PMCID: PMC1821076 DOI: 10.1053/jhep.2001.23438] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
To understand the physiological functions of exogenous hepatocyte growth factor (HGF) on normal adult animals, we delivered human HGF gene into mice by a hydrodynamics-based in vivo gene transfection approach using a naked plasmid vector. Systemic administration of naked plasmid containing HGF cDNA driven under cytomegalovirus promoter (pCMV-HGF) by rapid injection via the tail vein produced a remarkable level of human HGF protein in the circulation, beginning to appear at 4 hours and peaking at 12 hours following injection. Tissue distribution studies identified the liver as the organ with the highest level of transgene expression. Through weekly repeated injections of plasmid vector, we achieved sustained, long-term, high levels of exogenous HGF expression in mice for 8 weeks. Increases of more than 31% and 16% in liver and body weights were found, respectively, in the mice that received pCMV-HGF plasmid compared with that given the control vector for 8 weeks. Expression of exogenous HGF in vivo activated mitogen-activated protein kinases and induced proliferating cell nuclear antigen expression in normal adult liver and kidneys. These data suggest that systemic administration of naked plasmid vector is a convenient, safe, and highly efficient approach to introduce and maintain exogenous HGF gene expression in vivo in a controllable fashion. Our results also indicate that long-term expression of human HGF in mice markedly activates growth-related signal transduction events, promotes cell proliferation, and leads to liver and overall body growth in whole adult animals.
Collapse
Affiliation(s)
- Junwei Yang
- From the Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Shiping Chen
- Department of Cell Biology, Peking Union Medical College, Beijing, China
| | - Leaf Huang
- Center for Pharmacogenetics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA
| | - George K. Michalopoulos
- From the Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Youhua Liu
- From the Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
217
|
Mizuno S, Matsumoto K, Nakamura T. Hepatocyte growth factor suppresses interstitial fibrosis in a mouse model of obstructive nephropathy. Kidney Int 2001; 59:1304-14. [PMID: 11260391 DOI: 10.1046/j.1523-1755.2001.0590041304.x] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND As tubulointerstitial fibrosis (TIF) reflects the prognosis of patients with various chronic renal diseases, the pathogenesis of TIF has to be clarified. Transforming growth factor-beta (TGF-beta) is a key mediator for renal fibrosis. We reported that hepatocyte growth factor (HGF) prevents renal fibrosis in nephrotic mice. However, the function of HGF in chronic renal failure, except for nephrotic syndrome, remains to be determined. METHODS Using mice subjected to unilateral ureter-ligated obstruction (UUO), we investigated the roles of HGF in TIF, as induced by obstructive nephropathy. Pathophysiological changes in the kidney after UUO treatment were analyzed focusing on expressions of renal HGF and TGF-beta, TIF, tubular proliferation, and apoptosis. Neutralizing antibody against rodent HGF, or recombinant human HGF (rhHGF), was administrated to the UUO mice, and pathophysiological changes after neutralization or supplements of HGF were analyzed. RESULTS In this UUO model, TIF with tubular apoptosis became evident, and it was accompanied by a decrease in renal HGF expression and an increase in renal TGF-beta expression. Neutralization of endogenous HGF accelerated the progression of TIF, accompanied by increases in TGF-beta expression and tubular apoptosis as well as by decreases in tubular proliferation. In contrast, rhHGF attenuated TIF progression, and there were decreases in TGF-beta expression and tubular apoptosis, and an increase in tubular proliferation. CONCLUSIONS Endogenous as well as exogenous HGF attenuated the progression of the fibrosis caused by obstructive nephropathy in these mice. Thus, local reduction in HGF levels may account for TIF in chronic renal diseases.
Collapse
Affiliation(s)
- S Mizuno
- Division of Biochemistry, Department of Oncology, Biomedical Research Center, Osaka University Graduate School of Medicine, Yamadaoka, Japan
| | | | | |
Collapse
|
218
|
Gao C, Kennedy S, Ponder KP. Lipopolysaccharide potentiates the effect of hepatocyte growth factor upon replication in lung, thyroid, spleen, and colon in rats in vivo. Mol Ther 2001; 3:462-75. [PMID: 11319906 DOI: 10.1006/mthe.2001.0265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Induction of replication may potentiate in vivo gene therapy, as some viral vectors only transduce dividing cells. Hepatocyte growth factor (HGF) increases the percentage of replicating hepatocytes to 18-fold that in normal rats, and lipopolysaccharide (LPS) modestly potentiates this effect. In this study, the effect of iv HGF upon replication in other organs was determined. HGF at 10 mg/kg resulted in replication that was < or =3-fold that of normal rats in alveolar and proximal renal tubular cells. HGF alone had no effect upon replication of epithelial cells from the bronchi, thyroid, pancreas, or colon or upon cells from the muscle, pancreatic islets, spleen, blood vessels, or thymus. HGF and LPS at 5 mg/kg resulted in replication that was 9-fold that of normal rats in alveolar cells, 25-fold in bronchial epithelial cells, 4-fold in thyroid epithelial cells, 1.5-fold in the red pulp of the spleen, and 2-fold in colonic epithelial cells. The synergistic effect may be due to the fact that LPS upregulated the HGF receptor c-met in thyroid, spleen, and colon. We conclude that iv administration of HGF alone is relatively specific for inducing hepatocyte replication and would allow selective gene transfer into the liver.
Collapse
Affiliation(s)
- C Gao
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
219
|
Rabkin R, Fervenza F, Tsao T, Sibley R, Friedlaender M, Hsu F, Lassman C, Hausmann M, Huie P, Schwall RH. Hepatocyte growth factor receptor in acute tubular necrosis. J Am Soc Nephrol 2001; 12:531-540. [PMID: 11181801 DOI: 10.1681/asn.v123531] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
In acute tubular necrosis, there are early transient increases in circulating and local bioactive hepatocyte growth factor (HGF) levels and renal HGF receptor (c-MET) gene expression. It has therefore been suggested that endogenous HGF may play a role in initiating renal repair. To test this hypothesis, changes in the levels, activity, and anatomic distribution of c-MET protein were characterized in relation to the onset and localization of DNA synthesis in kidneys of rats with ischemia-induced acute tubular necrosis. Whole-kidney c-MET protein levels were significantly increased in the injured kidneys 12 h after injury and rose to a maximum after 1 d, exceeding the control values by sevenfold. Eight days after injury, c-MET levels, although decreasing, were still elevated above control values. An increase in the levels of activated c-MET, i.e., tyrosine-phosphorylated c-MET, was also evident as early as 12 h after injury. Histologic analyses demonstrated that the increase in c-MET immunoreactivity was most marked in the most severely damaged nephron segments in the outer medulla. In injured proximal tubules, the receptor was redistributed from an apical location to an intracellular location. DNA synthesis was increased in the injured kidneys, especially in the outer medulla, where the increase in c-MET protein levels was most prominent. The increase in DNA synthesis was first detected 12 h after the initial increase in activated c-MET levels. It is concluded that the early increases in the levels of c-MET protein and activated receptor support the hypothesis that HGF participates in the initiation of renal regeneration. In addition, the persistent elevation of c-Met protein levels suggests that prolonged and even late treatment with HGF may be of therapeutic value
Collapse
Affiliation(s)
- Ralph Rabkin
- Research Service Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California
- Department of Medicine, Stanford University, Palo Alto, California
| | - Fernando Fervenza
- Research Service Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California
- Department of Medicine, Stanford University, Palo Alto, California
| | - Tanny Tsao
- Research Service Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California
- Department of Medicine, Stanford University, Palo Alto, California
| | - Richard Sibley
- Department of Pathology, Stanford University, Palo Alto, California
| | - Michael Friedlaender
- Research Service Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California
- Department of Medicine, Stanford University, Palo Alto, California
| | - Fay Hsu
- Research Service Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California
- Department of Medicine, Stanford University, Palo Alto, California
| | - Charles Lassman
- Department of Pathology, Stanford University, Palo Alto, California
| | - Michael Hausmann
- Research Service Veterans Affairs Palo Alto Health Care System, Stanford University, Palo Alto, California
- Department of Medicine, Stanford University, Palo Alto, California
| | - Phil Huie
- Department of Pathology, Stanford University, Palo Alto, California
| | - Ralph H Schwall
- Department of Molecular Oncology, Genentech Inc., South San Francisco, California
| |
Collapse
|
220
|
Kobayashi Y, Nakamura N, Ishizaka T, Masuda K, Ohno K, Tsujimoto H. Molecular cloning of feline hepatocyte growth factor (HGF) cDNA. J Vet Med Sci 2001; 63:211-4. [PMID: 11258464 DOI: 10.1292/jvms.63.211] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hepatocyte growth factor (HGF) is a pleiotropic cytokine responsible for regeneration, development and maintenance of various organs, and growth, invasion and metastasis of tumor cells. A full-length feline HGF cDNA was cloned and sequenced by RT-PCR from cat liver. Feline HGF consists of 728 amino acid and contains alpha- and beta-chains encoded in a single open reading frame. The predicted amino acid sequence of feline HGF showed 93.2, 93.3 and 93.3% homology with those of human, mouse and rat HGF, respectively. The putative proteolytic processing site, all cysteine residues, and four potential glycosylation sites are conserved in all species. Therefore, feline HGF is expected to have a similar three-dimensional structure to human, mouse and rat HGF.
Collapse
Affiliation(s)
- Y Kobayashi
- Department of Veterinary Internal Medicine, Faculty of Agriculture, The University of Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
221
|
Dudkowska M, Stachurska A, Chmurzyska W, Grzelakowska-Sztabert B, Manteuffel-Cymborowska M. Cross-talk between steroid-receptor-mediated and cell-membrane-receptor-mediated signalling pathways results in the in vivo modulation of c-Met and ornithine decarboxylase gene expression in mouse kidney. Biochem J 2001; 353:317-23. [PMID: 11139396 PMCID: PMC1221574 DOI: 10.1042/0264-6021:3530317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The cross-talk in vivo between two signalling pathways activated by testosterone via intracellular androgen receptor, and induced by damage to renal tubules evoked by anti-folate [N(10)-propargyl-5,8-dideazafolic acid (CB 3717)] or folate is reported. We show that CB 3717/folate induces the expression of the hepatocyte growth factor (HGF)/c-Met signalling system in injured kidneys in which a significant, but transient, elevation of the HGF mRNA level occurs. It is followed by a severalfold increase in the c-Met transmembrane receptor message that persists for up to 24 h. The c-Met expression is also positively controlled by testosterone, which induces a significant increase in its mRNA level that is abolished by an anti-androgen, casodex. However, when testosterone and anti-folate/folate are administered sequentially, a substantial (3.5-4.0-fold) decrease in the increase of c-Met expression caused by CB 3717/folate alone occurs. Similarly, testosterone-induced ornithine decarboxylase (ODC) mRNA level and activity are decreased 2.8-7.7-fold when the androgen is applied together with CB 3717. Antagonism between these pathways is also visible under physiological conditions in the kidneys of male mice in which, owing to elevated endogenous testosterone levels, neither the ODC activity nor the mRNA level is induced by anti-folate/folate, whereas the c-Met message response to these drugs is significantly decreased. Our results document a substantial negative regulation of c-Met and ODC gene expression as a result of the cross-talk between testosterone-activated and HGF-activated pathways and suggest a sex-differentiated response to injury of mouse kidneys.
Collapse
Affiliation(s)
- M Dudkowska
- Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland.
| | | | | | | | | |
Collapse
|
222
|
Bulotta A, Di Cesare E, Ponte E, Falchi M, Sciacchitano S, Cucinotta D, Taruscio D, Di Mario U, Anastasi E. Increased c-met expression during ductal beta cell neogenesis in experimental autoimmune diabetes. Growth Factors 2001; 19:259-67. [PMID: 11811781 DOI: 10.3109/08977190109001091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
C-met immunoreactivity and its co-expression with duct-associated insulin were evaluated in pancreata of non-obese diabetic (NOD) and low-dose streptozotocin (Id-STZ) mice. Diabetic NOD and non-diabetic NOD at the age of 4-8, 15-22 and 30-41 weeks and Balb/c mice at the same age intervals were studied. Ld-STZ mice were studied at day 12 and 24 after STZ administration. A stronger ductal c-met immunoreactivity and a significantly higher number of c-met positive ducts were found in diabetic NOD vs both non-diabetic NOD and Balb/c mice of comparable age. In non-diabetic NOD, the ductal c-met immunoreactivity progressively increased with age and was significantly higher than controls. In 1d-STZ mice a significantly increased ductal c-met immunoreactivity was detected both at day 12 and 24 vs untreated mice. C-met positive ductal cells were also positive for insulin although insulin positive c-met negative ducts were present. This study showed an increased c-met expression and the co-expression of c-met and duct-associated insulin, in both NOD and 1d-STZ mice.
Collapse
Affiliation(s)
- A Bulotta
- Department of Clinical Sciences, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Kataoka H, Shimomura T, Kawaguchi T, Hamasuna R, Itoh H, Kitamura N, Miyazawa K, Koono M. Hepatocyte growth factor activator inhibitor type 1 is a specific cell surface binding protein of hepatocyte growth factor activator (HGFA) and regulates HGFA activity in the pericellular microenvironment. J Biol Chem 2000; 275:40453-62. [PMID: 11013244 DOI: 10.1074/jbc.m006412200] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hepatocyte growth factor activator (HGFA) is responsible for proteolytic activation of the precursor form of hepatocyte growth factor in injured tissues. To date, two specific inhibitors of HGFA have been identified, namely HGFA inhibitor type 1 (HAI-1) and type 2 (HAI-2)/placental bikunin (PB). Both inhibitors are first synthesized as integral membrane proteins having two Kunitz domains and a transmembrane domain, and are subsequently released from cell surface by shedding. Here we show that an active form of HGFA is specifically complexed with membrane-form HAI-1, but not with HAI-2/PB, on the surface of epithelial cells expressing both inhibitors. This binding required the enzyme activity of HGFA. The selective binding of HGFA to the cell surface HAI-1 was further confirmed in an engineered system using Chinese hamster ovary cells, in which only the cells expressing HAI-1 retained exogenous HGFA. The binding of HGFA to HAI-1 was reversible, and no irreversible modifications affecting the enzyme activity occurred during the binding. Importantly, HAI-1 and the HGFA.HAI-1 complex were quickly released from the cell surface by treatment with phorbol 12-myristate 13-acetate or interleukin 1beta accompanying the generation of 58-kDa fragments of HAI-1, which are less potent against HGFA, as well as significant recovery of HGFA activity in the culture supernatant. This regulated shedding was completely inhibited by BB3103, a synthetic zinc-metalloproteinase inhibitor. We conclude that HAI-1 is not only an inhibitor but also a specific acceptor of active HGFA, acting as a reservoir of this enzyme on the cell surface. The latter property appears to ensure the concentrated pericellular HGFA activity in certain cellular conditions, such as tissue injury and inflammation, via the up-regulated shedding of HGFA.HAI-1 complex. These findings shed light on a novel function of the integral membrane Kunitz-type inhibitor in the regulation of pericellular proteinase activity.
Collapse
Affiliation(s)
- H Kataoka
- Second Department of Pathology, Miyazaki Medical College, Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
224
|
Nakamura T, Mizuno S, Matsumoto K, Sawa Y, Matsuda H, Nakamura T. Myocardial protection from ischemia/reperfusion injury by endogenous and exogenous HGF. J Clin Invest 2000; 106:1511-9. [PMID: 11120758 PMCID: PMC387252 DOI: 10.1172/jci10226] [Citation(s) in RCA: 330] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Using a rat model of ischemia/reperfusion injury, we demonstrate here that HGF is cardioprotective due to its antiapoptotic effect on cardiomyocytes. Following transient myocardial ischemia and reperfusion, c-Met/HGF receptor expression rapidly increased in the ischemic myocardium, an event accompanied by a dramatic increase in plasma HGF levels in the infarcted rats. When endogenous HGF was neutralized with a specific antibody, the number of myocyte cell deaths increased markedly, the infarct area expanded, and the mortality increased to 50%, as compared with a control group in which there was no mortality. Plasma from the myocardial infarcted rats had cardioprotective effects on primary cultured cardiomyocytes, but these effects were significantly diminished by neutralizing HGF. In contrast, recombinant HGF administration reduced the size of infarct area and improved cardiac function by suppressing apoptosis in cardiomyocytes. HGF rapidly augmented Bcl-xL expression in injured cardiomyocytes both in vitro and in vivo. As apoptosis of cardiomyocytes is one of the major contributors to the pathogenesis in subjects with ischemia/reperfusion injury, prevention of apoptosis may prove to be a reasonable therapeutic strategy. Supplements of HGF, an endogenous cardioprotective factor, may be found clinically suitable in treating subjects with myocardial infarction.
Collapse
Affiliation(s)
- T Nakamura
- Division of Biochemistry, Department of Oncology, Biomedical Research Center B7, Osaka University Graduate School of Medicine, Suita, Japan.
| | | | | | | | | | | |
Collapse
|
225
|
Haug C, Schmid-Kotsas A, Zorn U, Bachem MG, Schuett S, Gruenert A, Rozdzinski E. Hepatocyte growth factor is upregulated by low-density lipoproteins and inhibits endothelin-1 release. Am J Physiol Heart Circ Physiol 2000; 279:H2865-71. [PMID: 11087242 DOI: 10.1152/ajpheart.2000.279.6.h2865] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Low-density lipoproteins (LDL) are known to cause endothelial injury and to promote the development of atherosclerotic lesions. This study demonstrates a significant concentration-dependent stimulatory effect of LDL on hepatocyte growth factor (HGF) synthesis (maximum release: 423 +/- 16% of control) and HGF receptor mRNA expression in cultured human coronary artery endothelial cells (HCAEC). HGF is a potent mitogen for endothelial cells but does not affect smooth muscle cell proliferation. In contrast, endothelin-1 (ET-1) acts as a mitogen on vascular smooth muscle cells and seems to be upregulated in coronary atherosclerosis. In this study, the basal ET-1 synthesis in HCAEC was concentration-dependently reduced by HGF (minimum: 54 +/- 3% of control). This inhibitory effect seems to be mediated via the tyrosine kinase activity of the HGF receptor c-met, since it was antagonized by the tyrosine kinase inhibitor lavendustin A. In addition, HGF also significantly reduced the LDL-stimulated ET-1 release. The LDL-induced upregulation of HGF synthesis in HCAEC and the inhibitory effect of HGF on ET-1 synthesis suggest a protective role of HGF in coronary atherosclerosis.
Collapse
Affiliation(s)
- C Haug
- Institute of Clinical Chemistry, University Hospital Ulm, D-89070 Ulm, Germany.
| | | | | | | | | | | | | |
Collapse
|
226
|
Cheng CH, Chen HC, Shu KH, Chen CH, Wu MJ, Lian JD. Increase of hepatocyte growth factor after renal transplantation. Transplant Proc 2000; 32:2070-2. [PMID: 11120071 DOI: 10.1016/s0041-1345(00)01562-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- C H Cheng
- Division of Nephrology, Taichung Veteran General Hospital, Taichung, Taiwan.
| | | | | | | | | | | |
Collapse
|
227
|
Liu Y, Rajur K, Tolbert E, Dworkin LD. Endogenous hepatocyte growth factor ameliorates chronic renal injury by activating matrix degradation pathways. Kidney Int 2000; 58:2028-2043. [PMID: 11044223 DOI: 10.1111/j.1523-1755.2000.00375.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Hepatocyte growth factor (HGF) has been shown to promote tubule repair and renal regeneration following acute injury; however, whether HGF also modulates the development and progression of chronic renal diseases that are characterized by progressive tissue fibrosis is uncertain. To examine this question, this study investigated the functional consequence of blocking endogenous HGF signaling in vivo in a model of chronic renal disease. The effects of HGF on the processes of matrix synthesis and degradation in cultured renal epithelial cells were also examined. METHODS The level of activity of the HGF/c-met axis was examined in rats following 5/6 nephrectomy at multiple time points. To determine the effects of HGF in modulating chronic renal injury, HGF action was blocked in remnant kidney rats using an anti-HGF antibody. The effects of HGF on extracellular matrix (ECM) synthesis and degradation were examined in renal epithelial cells by (35)S-methionine labeling, Western blotting, and zymographic analysis. RESULTS An increase in renal and systemic production of HGF coupled with an increase in renal c-met was observed in rats with remnant kidneys. When HGF action was blocked by the administration of an anti-HGF antibody, rats experienced a rapid decrease in glomerular filtration rate and increased renal fibrosis. Kidney sections from the antibody-treated rats displayed a marked increase in ECM accumulation and in alpha-smooth muscle actin-positive cells in both the interstitium and tubular epithelium. In vitro studies revealed that HGF reduced net ECM accumulation by human proximal tubule cells (HKC), and this effect was abolished by incubating cells with an anti-HGF antibody. HGF did not alter the ECM synthetic rate in HKC cells. Rather, it markedly increased collagenase such as matrix metalloproteinase-9 (MMP-9) protein expression, as evidenced by Western blotting and zymographic analysis. HGF also decreased the expression of tissue inhibitors of matrix metalloproteinase-1 (TIMP-1) and TIMP-2, the endogenous inhibitors of MMPs. CONCLUSION These results suggest that HGF is a potent antifibrogenic factor both in vitro and in vivo. Endogenous activation of HGF tends to preserve kidney structure and function in rats with chronic renal disease by activating matrix degradation pathways.
Collapse
Affiliation(s)
- Y Liu
- Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island, USA.
| | | | | | | |
Collapse
|
228
|
Yoshimura R, Kasai S, Wada S, Watanabe Y, Hase T, Chargui J, Nakatani T, Tanaka H, Yamamoto K, Kishimoto T. Hepatocyte growth factor: a sensitive indicator for the acute rejection of renal transplants. Transplant Proc 2000; 32:2073-4. [PMID: 11120072 DOI: 10.1016/s0041-1345(00)01573-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- R Yoshimura
- Department of Urology, Osaka City University School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Nigam ES, Lieberthal W. Acute renal failure. III. The role of growth factors in the process of renal regeneration and repair. Am J Physiol Renal Physiol 2000; 279:F3-F11. [PMID: 10894783 DOI: 10.1152/ajprenal.2000.279.1.f3] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
This review, which is the final installment in a series devoted to controversial issues in acute renal failure (ARF) (3, 47), will examine available information regarding the role of growth factors in ARF. In general, studies in this area have fallen into two broad categories: 1) those that have examined the renal expression of genes encoding growth factors or transcriptional factors associated with the growth response that is induced after ARF, and 2) those that have examined the efficacy of exogenously administered growth factors in accelerating recovery of renal function in experimental models of ARF. Despite the vast amount of information that has accumulated in these two areas of investigation, our understanding of the mechanisms involved in the process of regeneration and repair after ARF, and the role of growth factors in this response, remains rudimentary. This overview, contributed to by a number of experts in the field, is designed to summarize present knowledge and to highlight potentially fertile areas for future research in this area.
Collapse
Affiliation(s)
- e S Nigam
- Departments of Pediatrics and Medicine, University of California San Diego, La Jolla, California 92093-0693, USA
| | | |
Collapse
|
230
|
Matsumoto K, Mizuno S, Nakamura T. Hepatocyte growth factor in renal regeneration, renal disease and potential therapeutics. Curr Opin Nephrol Hypertens 2000; 9:395-402. [PMID: 10926176 DOI: 10.1097/00041552-200007000-00011] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Hepatocyte growth factor (HGF) has mitogenic, motogenic, morphogenic, and anti-apoptotic activities on renal cells and is a potential renotropin for renal protection and repair. In chronic renal failure/fibrosis, HGF in the kidney declines in a reciprocal manner to the increase in transforming growth factor-beta (TGF-beta). Neutralization of HGF by the antibody leads to acceleration of renal failure/fibrosis while HGF administration leads to remarkable attenuation, thus indicating the importance of HGF versus TGF-beta counterbalance in both pathogenesis and therapeutics in cases of chronic renal failure. HGF is being strongly considered for potential treatment of acute and chronic renal failure.
Collapse
Affiliation(s)
- K Matsumoto
- Department of Oncology, Biomedical Research Center, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | |
Collapse
|
231
|
Nagy P, Bisgaard HC, Schnur J, Thorgeirsson SS. Studies on hepatic gene expression in different liver regenerative models. Biochem Biophys Res Commun 2000; 272:591-5. [PMID: 10833457 DOI: 10.1006/bbrc.2000.2811] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have investigated the expression of several growth-related genes in the liver after partial hepatectomy in three experimental models: normal, Dexamethasone-pretreated, and hypophysectomized rats. Dexamethasone and hypophysectomy resulted in a delay in the peak of cell replication in 6 and 18 h, respectively, when compared to the normal animals. TGFalpha mRNA expression was shifted together with the DNA synthesis, but the expression of c-myc, c-fos, c-jun, HGF, TGFbeta1, IL1beta did not delay. This result suggests that liver-derived TGFalpha but not the other factors are important in the timing of the proliferative response after partial hepatectomy.
Collapse
Affiliation(s)
- P Nagy
- First Institute of Pathology and Experimental Cancer Research, Semmelweis University of Medicine, Budapest, Hungary.
| | | | | | | |
Collapse
|
232
|
Yasuda S, Noguchi T, Gohda M, Arai T, Tsutsui N, Matsuda T, Nonogi H. Single low-dose administration of human recombinant hepatocyte growth factor attenuates intimal hyperplasia in a balloon-injured rabbit iliac artery model. Circulation 2000; 101:2546-9. [PMID: 10831531 DOI: 10.1161/01.cir.101.21.2546] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Previous studies have shown that repeated systemic administration of human recombinant hepatocyte growth factor (hrHGF) in mg/kg levels modulates the wound-healing process in various diseases. Recently, HGF has been characterized as one of the most potent endothelial-cell-specific growth factors. We tested our hypothesis that local delivery of hrHGF, even at low microg/kg levels (> or =2 orders of magnitude lower than systemically administered doses), might attenuate neointimal hyperplasia in response to vascular injury via accelerated reendothelialization. METHODS AND RESULTS The iliac artery was denuded in 16 New Zealand White rabbits (3 kg), followed by administration, via a drug delivery catheter, of either hrHGF (10 microg; n = 11) or control vehicle (n=5) over 20 minutes. In pilot studies using this device, the drug permeated into the medial tissues, where it persisted for > or =24 hours. Four weeks after the local delivery of hrHGF, computer-assisted morphometric analysis revealed significant reduction in the intimal area (hrHGF, 0.37+/-0.21 versus control, 0.68+/-0.16 mm(2), mean +/- SD; P<0.05) but no change in the medial area (hrHGF, 1.03+/-0.21 versus control, 1.10+/-0.52 mm(2)). Scanning electron microscopy revealed extensive endothelialization with regular and confluent endothelial cell layer regeneration in the hrHGF-treated vessels. CONCLUSIONS Accelerated endothelialization after local delivery of hrHGF, a novel and potent endothelial cell mitogen, effectively attenuates neointimal proliferation even after single low-dose administration. This observation could have potential therapeutic implications in the prevention of restenosis after angioplasty.
Collapse
Affiliation(s)
- S Yasuda
- National Cardiovascular Center, Osaka, Japan.
| | | | | | | | | | | | | |
Collapse
|
233
|
Laping NJ, Olson BA, Ho T, Ziyadeh FN, Albrightson CR. Hepatocyte growth factor: a regulator of extracellular matrix genes in mouse mesangial cells. Biochem Pharmacol 2000; 59:847-53. [PMID: 10718343 DOI: 10.1016/s0006-2952(99)00398-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The potential role of hepatocyte growth factor (HGF) in regulating extracellular matrix in mouse mesangial cells (MMC) was evaluated. Functional HGF receptors were deed in MMC by HGF-induced extracellular acidification, a response that was inhibited by the HGF inhibitor HGF/NK2, a splice variant expressing the N-terminal domain through the second kringle domain HGF also increased fibronectin and collagen alpha1 (IV) mRNA levels in these cells; the increases were associated with a concentration-dependent increase in transcriptional activity of the collagen alpha1 (IV) gene. HGF also stimulated fibronectin and collagen alpha1 (IV) mRNA levels in primary rabbit proximal tubule epithelial cells To evaluate the potential consequences of chronic elevation of HGF on renal fuction, HGF was administered continuously for 18 days to normal and diabetic C57BLKS/J lepr(db) mice. In the diabetic mice, HGF reduced creatinine clearance and increased microalbuminuria, indicating that chronic exposure to HGF impairs renal function. Thus, chronically elevated HGF may contribute to the progression of chronic renal disease in diabetes by decreasing the glomerular filtration rate and possibly promoting the accumulation of extracellular matrix.
Collapse
Affiliation(s)
- N J Laping
- Department of Renal Pharmacology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA
| | | | | | | | | |
Collapse
|
234
|
Abstract
Can science discover some secrets of Greek mythology? In the case of Prometheus, we can now suppose that his amazing hepatic regeneration was caused by a peptide growth factor called hepatocyte growth factor (HGF). Increasing evidence indicates that HGF acts as a multifunctional cytokine on different cell types. This review addresses the molecular mechanisms that are responsible for the pleiotropic effects of HGF. HGF binds with high affinity to its specific tyrosine kinase receptor c-met, thereby stimulating not only cell proliferation and differentiation, but also cell migration and tumorigenesis. The three fundamental principles of medicine-prevention, diagnosis, and therapy-may be benefited by the rational use of HGF. In renal tubular cells, HGF induces mitogenic and morphogenetic responses. In animal models of toxic or ischemic acute renal failure, HGF acts in a renotropic and nephroprotective manner. HGF expression is rapidly up-regulated in the remnant kidney of nephrectomized rats, inducing compensatory growth. In a mouse model of chronic renal disease, HGF inhibits the progression of tubulointerstitial fibrosis and kidney dysfunction. Increased HGF mRNA transcripts were detected in mesenchymal and tubular epithelial cells of rejecting kidney. In transplanted patients, elevated HGF levels may indicate renal rejection. When HGF is considered as a therapeutic agent in human medicine, for example, to stimulate kidney regeneration after acute injury, strategies need to be developed to stimulate cell regeneration and differentiation without an induction of tumorigenesis.
Collapse
Affiliation(s)
- G A Vargas
- Department of Internal Medicine II, Division of Nephrology, University of Ulm, Germany
| | | | | |
Collapse
|
235
|
Abstract
It has been generally accepted that a catastrophic breakdown of regulated cellular homeostasis, known as necrosis, is the mode of cellular injury in various forms of acute renal failure. One of the major advances in our understanding of cell death has been the recognition that the pathways traditionally associated with apoptosis as described in the landmark study by Kerr, Wyllie, and Currie in 1972 maybe very critical in the form of cell injury associated with necrosis. The pathway that is followed by the cell varies with both nature and severity of insults and may evolve from an apoptotic to a necrotic form of cell death. It is also likely that there are some common pathways that are shared and regulated in the two modes of cell death. In this review, we first describe evidence for the role of apoptotic pathways in ischemic acute renal failure, and then consider the potential mechanisms that may participate in this model of acute renal tubular injury. We then summarize the current information of apoptotic pathways related to other common causes of acute renal failure including endotoxin-induced, toxic acute renal failure and transplant rejection. A better understanding of the mechanisms of apoptosis could lead to safer and more specific therapeutic interventions for acute renal failure.
Collapse
Affiliation(s)
- N Ueda
- Department of Medicine, University of Arkansas for Medical Sciences, and Central Arkansas Veterans Healthcare System, Little Rock 72205, USA
| | | | | |
Collapse
|
236
|
Nakano M, Yasunami Y, Maki T, Kodama S, Ikehara Y, Nakamura T, Tanaka M, Ikeda S. Hepatocyte growth factor is essential for amelioration of hyperglycemia in streptozotocin-induced diabetic mice receiving a marginal mass of intrahepatic islet grafts. Transplantation 2000; 69:214-21. [PMID: 10670629 DOI: 10.1097/00007890-200001270-00004] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND It is crucial for clinical islet transplantation to find a procedure to improve the success rate of insulin independence after islet transplantation. In the present study, we determined whether hepatocyte growth factor (HGF) has a favorable effect on amelioration of hyperglycemia in streptozotocin (STZ, 200 mg/kg)-induced diabetic mice (C57BL/6) receiving a marginal mass of intrahepatic islet isografts. METHODS Isolated syngeneic islets were transplanted into the liver of recipients. HGF with dextran sulfate (DS) was administered intraperitoneally once a day at day 0, 2, 4, 6, and 8 relative to islet transplantation. DS has been known to enhance the effect of HGF. RESULTS It was found that the number of 250 islets was a marginal mass as donor islets in this model, in which 2 out of 14 diabetic mice receiving 250 islets became normoglycemic by 90 days after transplantation. The treatment with HGF (100 microg) in conjunction with DS (200 microg) produced normoglycemia in all mice (n = 5). Morphological study as well as intraperitoneal glucose tolerance test revealed the beneficial effects of HGF. To our surprise, six out of nine mice receiving 250 islets and treated with DS alone became normoglycemic. Additional anti-HGF antibody treatment (100 microg, day -1, 0, 2, 4, 6, and 8) abolished the effects of DS, indicating that the effect by DS is mediated via the endogenous HGF. The effects of DS were not observed when the renal subcapsular space was the site of islet transplantation. There was a significant increase in plasma HGF levels in mice after the intrahepatic grafts but not the renal subcapsular one. CONCLUSIONS These findings demonstrate that HGF is essential for amelioration of hyperglycemia in STZ-induced diabetic mice when a marginal mass of islets was grafted into the liver. As the liver is the site of clinical islet transplantation and the inability to achieve insulin independence after transplantation is a major obstacle for successful transplantation, HGF may facilitate to overcome such an important issue for clinical islet transplantation.
Collapse
Affiliation(s)
- M Nakano
- Department of Surgery I, Fukuoka University School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
237
|
Rampino T, Arbustini E, Gregorini M, Guallini P, Libetta C, Maggio M, Ranghino A, Silini E, Soccio G, Dal Canton A. Hemodialysis prevents liver disease caused by hepatitis C virus: role of hepatocyte growth factor. Kidney Int 1999; 56:2286-91. [PMID: 10594807 DOI: 10.1046/j.1523-1755.1999.00791.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
UNLABELLED Hemodialysis prevents liver disease caused by hepatitis C virus: Role of hepatocyte growth factor. BACKGROUND Hemodialysis increases markedly the serum levels of hepatocyte growth factor (HGF) so that regular dialysis treatment (RDT) mimics the regular administration of HGF as a drug. Therefore, we have studied the effects of dialysis-associated HGF production on the severity of liver damage caused by hepatitis C virus (HCV). METHODS Biochemical tests of liver function and liver biopsy were performed in 10 patients on RDT and in 11 patients without renal disease (WRD) converted to anti-HCV serum-positive test for the same time (48 +/- 4 months). The HGF serum concentration was measured by enzyme immunoassay. In patients on RDT, HGF was measured just before starting a dialysis session (T0), at 15 and 240 minutes of dialysis (T15 and T240), and 24 hours later (T24 hr). RESULTS Serum HGF was similar in WRD (average 0.17 ng/ml) as in RDT at T0 (0.25 ng/ml). In RDT serum HGF increased markedly at T15 and T240 (5.51 and 2.67 ng/ml, respectively, P < 0. 001 vs. WRD and T0) and was still higher than baseline at T24 hr (0. 41 ng/ml, P < 0.05). Both grade of necroinflammatory activity and stage of fibrosis were significantly lower in RDT than in WRD (both, P < 0.001). The number of apoptotic hepatocytes was also significantly reduced in patients on RDT compared with patients WRD. CONCLUSION These results show that HCV-related liver disease is more benign in patients on RDT. The phenomenon may depend on the marked and prolonged HGF release caused by dialysis.
Collapse
Affiliation(s)
- T Rampino
- Unità di Nefrologia e Dialisi and Servizio di Anatomia Patologica, Università ed IRCCS Ospedale San Matteo, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Comoglio PM, Tamagnone L, Boccaccio C. Plasminogen-related growth factor and semaphorin receptors: a gene superfamily controlling invasive growth. Exp Cell Res 1999; 253:88-99. [PMID: 10579914 DOI: 10.1006/excr.1999.4684] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Plasminogen-related growth factors (PRGFs), also known as "scatter factors," trigger a unique biological program leading to "invasive growth." This is a result of the integration of apparently independent biological responses including cell proliferation, cell survival, cell motility, invasion of extracellular matrices, and induction of cell polarity. Under physiological conditions, the coordinated execution of the underlying genetic programs leads to the formation of tubular structures by epithelial organs (the so-called branching morphogenesis). PRGF receptors are tyrosine kinases, encoded by a family of oncogenes: MET and RON. They feature unique signal transduction properties as their cytoplasmic tails contain a two-tyrosine multifunctional docking site that binds multiple SH2-containing intracellular signal transducers. Invasive growth results from the concomitant activation of Ras (growth), phosphatidylinositol 3-kinase ("scattering"), and signal transducer and activator of transcription (cell polarity and morphogenesis). We recently identified a new human gene family, encoding large transmembrane proteins, sex/plexins, sharing homologies with Met. These molecules are receptors for semaphorins, involved in axon guidance and cell-cell repulsion, a process reminiscent of scattering and invasive growth. Deregulated activation of PRGF or semaphorin ligands or receptors, by mutation or overexpression, confers to cancer cells invasive and metastatic properties.
Collapse
Affiliation(s)
- P M Comoglio
- Institute for Cancer Research and Treatment, University of Turin School of Medicine, Candiolo, 10060, Italy.
| | | | | |
Collapse
|
239
|
Yoshimura R, Kasai S, Watanabe Y, Hase T, Wada S, Yoshimura N, Kishimoto T. Hepatocyte growth factor in renal transplantation. Transplant Proc 1999; 31:2711-2. [PMID: 10578261 DOI: 10.1016/s0041-1345(99)00536-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- R Yoshimura
- Department of Urology, Osaka City University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
240
|
Nagahori T, Dohi M, Matsumoto K, Saitoh K, Honda ZI, Nakamura T, Yamamoto K. Interferon-gamma upregulates the c-Met/hepatocyte growth factor receptor expression in alveolar epithelial cells. Am J Respir Cell Mol Biol 1999; 21:490-7. [PMID: 10502559 DOI: 10.1165/ajrcmb.21.4.3614] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In the repair process after lung injury, the regeneration of alveolar epithelial cells plays an important role by covering the damaged alveolar wall and preventing the activated fibroblasts from invading the intra- alveolar spaces. Hepatocyte growth factor (HGF) is a potent mitogen for alveolar epithelial cells and has been reported to be capable of repressing the fibrosing process by connecting to the c-Met/HGF receptor on alveolar epithelial cells. However, it has been reported that the c-Met expression was downregulated in an acute phase of lung injury, which may limit the effect of HGF for therapeutic use. In the present study we observed that interferon (IFN)-gamma upregulates the c-Met messenger RNA (mRNA) and protein expression in A549 alveolar epithelial cells. We analyzed the mechanism of this upregulation and found that IFN-gamma enhances the transcription of the c-met proto-oncogene, and that it does not prolong the stability of the c-Met mRNA. HGF is known to act as a motogen as well as a mitogen for epithelial cells. We also found that the migratory activity of A549 cells induced by HGF is strongly enhanced by preincubation with IFN-gamma. Finally, we administered recombinant IFN-gamma to C57BL/6 mice and confirmed that this upregulation is also observed in vivo. These results suggest that the combination of HGF and IFN-gamma could be a new therapeutic approach for fibrosing pulmonary diseases.
Collapse
Affiliation(s)
- T Nagahori
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
241
|
Liu Y. Hepatocyte growth factor promotes renal epithelial cell survival by dual mechanisms. Am J Physiol Renal Physiol 1999; 277:F624-F633. [PMID: 10516287 DOI: 10.1152/ajprenal.1999.277.4.f624] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hepatocyte growth factor (HGF) has been shown to protect renal epithelial cells against apoptosis. To define the mechanism by which HGF inhibits apoptosis, we investigated the effect of HGF on the phosphorylation and expression of the Bcl-2 family proteins. Using a human proximal tubular epithelial cell (HKC) line as a model, we demonstrated that constitutive expression of HGF conveyed marked resistance to apoptotic death induced by serum withdrawal. HGF induced rapid phosphorylation of Akt in HKC cells, which was immediately followed by phosphorylation and resultant inactivation of Bad, a pro-apoptotic member of the Bcl-2 family. Pretreatment of the HKC cells with 10 nM wortmannin completely abolished HGF-induced phosphorylation of Akt and Bad, suggesting that this pathway is dependent on phosphoinositide (PI) 3-kinase. Overexpression of Bad increased apoptotic death in wild-type HKC cells but not in HGF-producing H4 cells. Immunoblotting confirmed that the Bad protein over-expressed in H4 cells was fully phosphorylated at both Ser(112) and Ser(136) sites. Prolonged incubation of HKC cells with HGF also dramatically induced expression of Bcl-xL, an anti-apoptotic member of the Bcl-2 family. These results suggest that the anti-apoptotic effect of HGF in renal epithelial cells is mediated by dual mechanisms involving two distinct Bcl-2 family proteins. HGF triggers Bad phosphorylation via the PI 3-kinase/Akt pathway, thereby inactivating this pro-apoptotic protein, while simultaneously inducing expression of anti-apoptotic Bcl-xL.
Collapse
Affiliation(s)
- Y Liu
- Department of Medicine, Rhode Island Hospital, Providence, Rhode Island 02903, USA
| |
Collapse
|
242
|
Koochekpour S, Jeffers M, Wang PH, Gong C, Taylor GA, Roessler LM, Stearman R, Vasselli JR, Stetler-Stevenson WG, Kaelin WG, Linehan WM, Klausner RD, Gnarra JR, Vande Woude GF. The von Hippel-Lindau tumor suppressor gene inhibits hepatocyte growth factor/scatter factor-induced invasion and branching morphogenesis in renal carcinoma cells. Mol Cell Biol 1999; 19:5902-12. [PMID: 10454537 PMCID: PMC84441 DOI: 10.1128/mcb.19.9.5902] [Citation(s) in RCA: 154] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/1999] [Accepted: 06/03/1999] [Indexed: 11/20/2022] Open
Abstract
Loss of function in the von Hippel-Lindau (VHL) tumor suppressor gene occurs in familial and most sporadic renal cell carcinomas (RCCs). VHL has been linked to the regulation of cell cycle cessation (G(0)) and to control of expression of various mRNAs such as for vascular endothelial growth factor. RCC cells express the Met receptor tyrosine kinase, and Met mediates invasion and branching morphogenesis in many cell types in response to hepatocyte growth factor/scatter factor (HGF/SF). We examined the HGF/SF responsiveness of RCC cells containing endogenous mutated (mut) forms of the VHL protein (VHL-negative RCC) with that of isogenic cells expressing exogenous wild-type (wt) VHL (VHL-positive RCC). We found that VHL-negative 786-0 and UOK-101 RCC cells were highly invasive through growth factor-reduced (GFR) Matrigel-coated filters and exhibited an extensive branching morphogenesis phenotype in response to HGF/SF in the three-dimensional (3D) GFR Matrigel cultures. In contrast, the phenotypes of A498 VHL-negative RCC cells were weaker, and isogenic RCC cells ectopically expressing wt VHL did not respond at all. We found that all VHL-negative RCC cells expressed reduced levels of tissue inhibitor of metalloproteinase 2 (TIMP-2) relative to the wt VHL-positive cells, implicating VHL in the regulation of this molecule. However, consistent with the more invasive phenotype of the 786-0 and UOK-101 VHL-negative RCC cells, the levels of TIMP-1 and TIMP-2 were reduced and levels of the matrix metalloproteinases 2 and 9 were elevated compared to the noninvasive VHL-positive RCC cells. Moreover, recombinant TIMPs completely blocked HGF/SF-mediated branching morphogenesis, while neutralizing antibodies to the TIMPs stimulated HGF/SF-mediated invasion in vitro. Thus, the loss of the VHL tumor suppressor gene is central to changes that control tissue invasiveness, and a more invasive phenotype requires additional genetic changes seen in some but not all RCC lines. These studies also demonstrate a synergy between the loss of VHL function and Met signaling.
Collapse
Affiliation(s)
- S Koochekpour
- ABL Basic Research Program, NCI Frederick Cancer Research and Development Center, Frederick, Maryland 21702, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Broten J, Michalopoulos G, Petersen B, Cruise J. Adrenergic stimulation of hepatocyte growth factor expression. Biochem Biophys Res Commun 1999; 262:76-79. [PMID: 10448071 DOI: 10.1006/bbrc.1999.1183] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatocyte growth factor (HGF), a potent mitogen, is released into plasma at increased levels following injury to certain tissues, including the liver. Early increases in plasma HGF are not due to a release from the injured liver, but rather from distal organs, particularly the lung. We have investigated the ability of norepinephrine (NE), which rises rapidly in plasma after liver resection, to trigger elevated production of HGF in MRC-5 human embryonic lung fibroblasts. Levels of HGF released to culture media and of HGF mRNA increased when cultures were exposed to NE, or to other adrenergic agonists. While stimulation of either beta- or alpha(1)-adrenergic receptors increased HGF expression, responses to NE appear to be mediated primarily via beta receptors. Since NE has already been shown to act as a comitogen with HGF, our findings suggest that adrenergic hormones may act both to induce production of HGF at distal sites, and to enhance the response to HGF at target tissues.
Collapse
Affiliation(s)
- J Broten
- Department of Biology, University of St. Thomas, St. Paul, Minnesota, 55105, USA
| | | | | | | |
Collapse
|
244
|
Matsumoto K, Morishita R, Moriguchi A, Tomita N, Yo Y, Nishii T, Nakamura T, Higaki J, Ogihara T. Prevention of renal damage by angiotensin II blockade, accompanied by increased renal hepatocyte growth factor in experimental hypertensive rats. Hypertension 1999; 34:279-84. [PMID: 10454454 DOI: 10.1161/01.hyp.34.2.279] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hepatocyte growth factor (HGF) is a unique growth factor that has many protective functions against renal damage. Our previous study demonstrated that HGF stimulated the growth of endothelial and epithelial cells without the replication of mesangial cells. Moreover, angiotensin (Ang) II significantly decreased local HGF production in mesangial cells. Therefore, we examined the effects of Ang II blockade on renal HGF expression and renal damage in experimental hypertensive rats. An angiotensin-converting enzyme inhibitor (cilazapril; 10 mg. kg(-1). d(-1)), an Ang II type 1 receptor antagonist (E-4177; 30 mg. kg(-1). d(-1)), hydralazine (8 mg. kg(-1). d(-1)), and vehicle were administered to 16-week-old stroke-prone spontaneously hypertensive rats (SHR-SP) for 3 weeks. Renal damage was evaluated with a computer analysis system, and renal HGF mRNA was measured by Northern blot analysis. Blood pressure of SHR-SP was significantly decreased by all drug treatments compared with vehicle. Moreover, cilazapril, E-4177, and hydralazine significantly decreased the thickening and necrosis of blood vessels compared with vehicle. Similarly, degeneration and necrosis of glomeruli were also markedly improved by cilazapril and E-4177 (P<0.01). We next examined the effects of Ang II blockade on renal HGF expression in SHR-SP. Renal HGF mRNA was markedly decreased in SHR-SP compared with Wistar-Kyoto rats, although Ang II blockade by cilazapril and E-4177 but not hydralazine significantly increased renal HGF mRNA in SHR-SP. Ang II blockade significantly increased renal HGF (a protective growth factor for tubular epithelial cells); thus, we examined tubular histological appearance. Degeneration and necrosis of tubules were significantly improved by cilazapril and E-4177 treatment (P<0.01). In addition, cell infiltration into the glomeruli and hemorrhage were also significantly reduced in SHR-SP treated with cilazapril or E-4177. The present data demonstrated the prevention of renal damage by Ang II blockade in SHR-SP, which was accompanied by a significant increase in renal HGF mRNA. Given the strong mitogenic activity and antiapoptotic actions of HGF on endothelial and epithelial cells, we believe that increased local HGF production by the blockade of Ang II may improve renal function in hypertension.
Collapse
Affiliation(s)
- K Matsumoto
- Department of Geriatric Medicine, Division of Gene Therapy Science, Osaka University Medical School, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Bowes RC, Lightfoot RT, Van De Water B, Stevens JL. Hepatocyte growth factor induces tubulogenesis of primary renal proximal tubular epithelial cells. J Cell Physiol 1999; 180:81-90. [PMID: 10362020 DOI: 10.1002/(sici)1097-4652(199907)180:1<81::aid-jcp9>3.0.co;2-j] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Hepatocyte growth factor (HGF)-induced tubulogenesis has been demonstrated with renal epithelial cell lines grown in collagen gels but not with primary cultured renal proximal tubular epithelial cells (RPTEs). We show that HGF selectively induces proliferation and branching morphogenesis of primary cultured rat RPTEs. Additional growth factors including fibroblast growth factor (FGF)-1, epidermal growth factor (EGF), FGF-7, or insulin-like growth factor-1 (IGF-1) did not selectively induce tubulogenesis. However, when administered in combination, these factors initiated branching morphogenesis comparable to HGF alone and greatly augmented HGF-induced proliferation and branching. Microscopic analysis revealed that branching RPTEs were undergoing tubulogenesis and formed a polarized epithelium. TGF-beta1 blocked HGF- or growth factor cocktail (GFC; HGF, FGF-1, EGF, IGF-1)-induced proliferation and branching morphogenesis. Adding TGF-beta1 after GFC-induced tubulogenesis had occurred caused a progressive regression of the tubular structures, a response associated with an increase in apoptosis of the RPTEs. Primary cultured RPTEs are capable of undergoing HGF-induced tubulogenesis. Unlike cell lines, combinations of growth factors differentially augment the response.
Collapse
Affiliation(s)
- R C Bowes
- Campbell University School of Pharmacy, Department of Pharmaceutical Sciences, Buies Creek, North Carolina 27506, USA.
| | | | | | | |
Collapse
|
246
|
Kosai K, Matsumoto K, Funakoshi H, Nakamura T. Hepatocyte growth factor prevents endotoxin-induced lethal hepatic failure in mice. Hepatology 1999; 30:151-9. [PMID: 10385651 DOI: 10.1002/hep.510300102] [Citation(s) in RCA: 164] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sepsis and endotoxemia are involved in the development of fulminant hepatic failure, the prognosis of which is extremely poor and the mortality is high, with no available effective therapy. Here, we report that hepatocyte growth factor (HGF) exerts potent antiapoptotic effects in vivo and effectively prevents endotoxin-induced fulminant hepatic failure in mice. The animals were intraperitoneally injected three times with 120 micrograms human recombinant HGF or saline 6 hours and 30 minutes before and 3 hours after an intraperitoneal injection of lipopolysaccharide (LPS) and D-galactosamine (GalN). Administration of LPS + GalN, without HGF, rapidly led to massive hepatocyte apoptosis and severe liver injury, and all mice died of hepatic failure within 8 hours. In contrast, administration of human recombinant HGF strongly suppressed extensive progress of hepatocyte apoptosis and the liver injury induced by LPS + GalN, and 75% of the HGF-treated mice survived. Moreover, HGF strongly induced Bcl-xL expression and blocked apoptotic signal transduction upstream of CPP32 (caspase-3) in the liver, thereby leading to inhibition of massive hepatocyte apoptosis. We suggest that HGF may well have the potential to prevent fulminant hepatic failure, at least through its potent antiapoptotic action.
Collapse
Affiliation(s)
- K Kosai
- Division of Biochemistry, Department of Oncology, Biomedical Research Center, Osaka University Medical School, Suita, Osaka, Japan
| | | | | | | |
Collapse
|
247
|
Ueda H, Sawa Y, Matsumoto K, Kitagawa-Sakakida S, Kawahira Y, Nakamura T, Kaneda Y, Matsuda H. Gene transfection of hepatocyte growth factor attenuates reperfusion injury in the heart. Ann Thorac Surg 1999; 67:1726-31. [PMID: 10391282 DOI: 10.1016/s0003-4975(99)00279-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Hepatocyte growth factor (HGF), a ligand for the c-Met receptor tyrosine kinase, plays a role as organotrophic factor for regeneration of various organs. HGF has an angiogenic activity and exhibits a potent antiapoptotic activity in several types of cells. Although HGF and the c-Met/HGF receptor are expressed in the heart, the role of HGF in the heart has remained unknown. METHODS After we analyzed changes in expression of endogenous HGF and c-Met mRNA levels in the rat left ventricle after myocardial infarction, the human HGF gene in hemagglutinating virus of Japan (HVJ)-liposome was transfected into the normal whole rat heart. Three days after transfection, the heart was subjected to global warm ischemia and subsequent reperfusion, followed by assessment of its cardiac functions. RESULTS Both HGF and c-Met/HGF receptor mRNAs were expressed in adult rat heart, and c-Met/HGF receptor mRNA was upregulated in response to myocardial infarction. HGF-transfected heart showed significant increase of human HGF protein level in the heart. Cardiac functions in terms of the left ventricular developed pressure, maximum dp/dt, and pressure rate product in hearts with HGF gene transfection were significantly superior to those in control hearts. In addition, leakage of creatine phosphokinase in the coronary artery effluent in hearts with HGF gene transfection was significantly lower than that in control hearts. CONCLUSIONS These data indicated that both HGF and c-Met/HGF receptor mRNAs were upregulated in response to myocardial ischemic injury, and that HGF is likely to have a cytoprotective effect on cardiac tissue, presumably through the c-Met/HGF receptor.
Collapse
Affiliation(s)
- H Ueda
- First Department of Surgery, Biomedical Research Center, Osaka University Medical School, Suita, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
248
|
Kataoka H, Suganuma T, Shimomura T, Itoh H, Kitamura N, Nabeshima K, Koono M. Distribution of hepatocyte growth factor activator inhibitor type 1 (HAI-1) in human tissues. Cellular surface localization of HAI-1 in simple columnar epithelium and its modulated expression in injured and regenerative tissues. J Histochem Cytochem 1999; 47:673-82. [PMID: 10219059 DOI: 10.1177/002215549904700509] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We used a specific monoclonal antibody to human hepatocyte growth factor activator inhibitor type 1 (HAI-1) in immunohistochemical procedures to determine the distribution and localization of HAI-1 in human tissues. In normal adult tissues, HAI-1 was predominantly expressed in the simple columnar epithelium of the ducts, tubules, and mucosal surface of various organs. In all cases, HAI-1 was localized predominantly on the cellular lateral (or basolateral) surface. By contrast, hepatocytes, acinar cells, endocrine cells, stromal mesenchymal cells, and inflammatory cells were hardly stainable with the antibody, and stratified squamous epithelium showed only faint immunoreactivity on the surface of cells of the basal layer. In the gastrointestinal tract, the surface epithelium was strongly stained. RNA blot analysis confirmed the presence of specific mRNA transcript in the gastrointestinal mucosa, and in situ hybridization revealed that HAI-1 mRNA showed a similar cellular distribution pattern. Although HAI-1 was not expressed in normal hepatocytes, strong immunoreactivity was observed on the epithelium of pseudo-bile ducts and on the surface of scattered hepatocytes in fulminant hepatitis. The enhanced expression was also noted in regenerating tubule epithelial cells of the kidney after infarction. We conclude that HAI-1 is preferentially expressed in the simple columnar epithelium of the mucosal surface and duct, that the predominant localization of HAI-1 is the cell surface, and that the expression of HAI-1 can be modulated by tissue injury and regeneration.
Collapse
Affiliation(s)
- H Kataoka
- Second Department of Pathology, Miyazaki Medical College, Miyazaki, Japan
| | | | | | | | | | | | | |
Collapse
|
249
|
Nishimura M, Ikeda T, Ushiyama M, Nanbu A, Kinoshita S, Yoshimura M. Increased vitreous concentrations of human hepatocyte growth factor in proliferative diabetic retinopathy. J Clin Endocrinol Metab 1999; 84:659-62. [PMID: 10022434 DOI: 10.1210/jcem.84.2.5434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human hepatocyte GF (hHGF) has strong neoangiogenesis activity. The present study was designed to investigate the possible involvement of hHGF in neovascularization in proliferative diabetic retinopathy (PDR) by measuring vitreous hHGF concentrations. The mean vitreous hHGF concentration was higher in subjects with PDR (5.70 +/- 0.68 ng/mL, n = 33) than in nondiabetic control subjects (1.50 +/- 0.20 ng/mL, n = 18, P < 0.01), nondiabetic subjects with proliferative vitreoretinopathy (3.31 +/- 0.57 ng//mL, n = 10, P < 0.05), or diabetic subjects without PDR (1.29 +/- 0.28 ng/mL, n = 8, P < 0.01). PDR subjects with neovascularization of iris, which suggests advanced retinal ischemia, showed a higher mean vitreous hHGF concentration than those without iridal neovascularization [7.33 +/- 1.16 ng/mL (n = 14) vs. 4.49 +/- 0.72 ng/mL (n = 19), P < 0.05]. The mean vitreous hHGF concentration was higher in PDR subjects with retinal neovascularization at the optic disc than in those with neovascularization elsewhere [7.3 +/- 1.1 ng/mL (n = 15) vs. 4.4 +/- 0.7 ng/mL (n = 18), P < 0.05]. Our results indicate that vitreous hHGF may play a role in retinal neovascularization in PDR.
Collapse
Affiliation(s)
- M Nishimura
- Department of Clinical and Laboratory Medicine, Kyoto Prefectural University of Medicine, Japan.
| | | | | | | | | | | |
Collapse
|
250
|
Liu Y, Tolbert EM, Lin L, Thursby MA, Sun AM, Nakamura T, Dworkin LD. Up-regulation of hepatocyte growth factor receptor: an amplification and targeting mechanism for hepatocyte growth factor action in acute renal failure. Kidney Int 1999; 55:442-453. [PMID: 9987069 DOI: 10.1046/j.1523-1755.1999.00267.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Hepatocyte growth factor (HGF) and its c-met receptor comprise a signaling system that has been implicated in tissue repair and regeneration. HGF action is specifically targeted to the damaged organ following injury; however, the mechanism underlying this important targeting process remains to be elucidated. We reasoned that induction of c-met expression might be a critical factor in determining the site specificity of this receptor-ligand system. To test this hypothesis, we examined changes in activity of the HGF/c-met system in the folic acid model of acute tubular injury and repair. METHODS Tissue HGF and c-met mRNA levels were detected by RNase protection assay and Northern blot analysis following acute renal injury induced by a single injection of folic acid. HGF and c-met proteins were examined by a specific enzyme immunoassay and Western blotting, respectively. C-met expression and trans-activation were investigated by exposing renal epithelial mIMCD-3 cells to various cytokines in vitro. RESULTS Extremely rapid induction of renal HGF and c-met mRNA was observed beginning one hour following injection of folic acid. Circulating plasma HGF protein level rose dramatically (approximately 16-fold), peaking first at two hours and again at 24 hours following injection. Despite elevated HGF mRNA in the kidney, total kidney HGF protein actually decreased significantly at 24 hours following injury. On the other hand, both c-met mRNA and c-met protein were markedly increased in the kidney, where active renal tubule repair and regeneration take place. In vitro studies suggested that increased levels of HGF, as well as other cytokines, might account for enhanced c-met expression in renal tubular epithelial cells. Pretreatment of the cells with actinomycin D totally blocked c-met induction, suggesting that induced c-met expression occurs primarily at the transcriptional level. Using a cloned region of the c-met promoter coupled to a reporter gene, we demonstrated that HGF directly stimulated c-met promoter transactivation in renal epithelial cells. CONCLUSION These results suggest that local up-regulation of c-met transcription in the kidney is crucial to renal tubule repair and regeneration, not only because it increases overall activity of this receptor-ligand system, but also as a mechanism targeting HGF action specifically to renal epithelia.
Collapse
Affiliation(s)
- Y Liu
- Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island, USA.
| | | | | | | | | | | | | |
Collapse
|