1
|
Miyabe Y, Sekiya S, Sugiura N, Oka M, Karasawa K, Moriyama T, Nitta K, Shimizu T. Renal subcapsular transplantation of hepatocyte growth factor-producing mesothelial cell sheets improves ischemia-reperfusion injury. Am J Physiol Renal Physiol 2019; 317:F229-F239. [DOI: 10.1152/ajprenal.00601.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a clinically important cause of acute kidney injury leading to chronic kidney disease. Furthermore, IRI in renal transplantation still remains a risk factor for delayed graft function. Previous studies on IRI have had some limitations, and few of the studied therapies have been clinically applicable. Therefore, a new method for treating renal IRI is needed. We examined the effects of human mesothelial cell (MC) sheets and hepatocyte growth factor (HGF)-transgenic MC (tg MC) sheets transplanted under the renal capsule in an IRI rat model and compared these two treatments with the intravenous administration of HGF protein and no treatment through serum, histological, and mRNA analyses over 28 days. MC sheets and HGF-tg MC sheets produced HGF protein and significantly improved acute renal dysfunction, acute tubular necrosis, and survival rate. The improvement in necrosis was likely due to the cell sheets promoting the migration and proliferation of renal tubular cells, as observed in vitro. Expression of α-smooth muscle actin at day 14 and renal fibrosis at day 28 after IRI were significantly suppressed in MC sheet and HGF-tg MC sheet treatment groups compared with the other groups, and these effects tended to be reinforced by the HGF-tg MC sheets. These results suggest that the cell sheets locally and continuously affect renal paracrine factors, such as HGF, and support recovery from acute tubular necrosis and improvement of renal fibrosis in chronic disease.
Collapse
Affiliation(s)
- Yoei Miyabe
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Medicine, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Sachiko Sekiya
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| | - Naoko Sugiura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Medicine, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Masatoshi Oka
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Medicine, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kazunori Karasawa
- Department of Medicine, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Takahito Moriyama
- Department of Medicine, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kosaku Nitta
- Department of Medicine, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
2
|
Wang Z, Fei S, Suo C, Han Z, Tao J, Xu Z, Zhao C, Tan R, Gu M. Antifibrotic Effects of Hepatocyte Growth Factor on Endothelial-to-Mesenchymal Transition via Transforming Growth Factor-Beta1 (TGF-β1)/Smad and Akt/mTOR/P70S6K Signaling Pathways. Ann Transplant 2018; 23:1-10. [PMID: 29292365 PMCID: PMC6248046 DOI: 10.12659/aot.906700] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background The related mechanisms involved in allograft interstitial fibrosis and chronic allograft dysfunction (CAD), following renal transplant, remain largely unknown. Here, we explored the role of hepatocyte growth factor (HGF) treatment on the endothelial-to-mesenchymal transition (EndMT) as a new way to target and prevent kidney fibrosis and improve outcomes for renal transplant recipients. Method/Material We extracted proteins and mRNAs from human umbilical vein endothelial cells (HUVECs) and human renal glomerular endothelial cells (HRGECs) treated with transforming growth factor-beta1 (TGF-β1) and/or varying doses of HGF, and assessed the effect of HGF on the EndMT using western blotting, qRT-PCR, and ELISA assays. We utilized cell motility and migration assays to evaluate cell movement, and applied western blotting to assess the mechanism by which TGF-β1 induced the EndMT. Results HGF significantly attenuated the development of TGF-β1-induced EndMT in a concentration-dependent way, and weakened the abilities of motility and migration of both HUVECs and HRGECs. Moreover, our results reveal that the antifibrotic effect of HGF on the EndMT was associated with the TGF-β/Smad and Akt/mTOR/p70S6K signaling pathways. Conclusions Our study suggests that HGF treatment significantly attenuates the development of EndMT induced by TGF-β1 via the TGFβ/Smad and Akt/mTOR/P70S6K signaling, which provides novel insights into the prevention and treatment of interstitial fibrosis and CAD following renal transplant.
Collapse
Affiliation(s)
- Zijie Wang
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Shuang Fei
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Chuanjian Suo
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Zhijian Han
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Jun Tao
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Zhen Xu
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Chunchun Zhao
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Min Gu
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
3
|
Wang HY, Zhang C, Xiao QF, Dou HC, Chen Y, Gu CM, Cui MJ. Hepatocyte growth factor inhibits tubular epithelial‑myofibroblast transdifferentiation by suppression of angiotensin II via the JAK2/STAT3 signaling pathway. Mol Med Rep 2017; 15:2737-2743. [PMID: 28447719 DOI: 10.3892/mmr.2017.6301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/01/2017] [Indexed: 11/06/2022] Open
Abstract
Tubular epithelial‑myofibroblast transdifferentiation (TEMT) is important in the development of chronic renal failure. The present study investigated whether hepatocyte growth factor (HGF) inhibits TEMT, and whether this function may be associated with the inhibition of angiotensin II (AngII) and the Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway. Human HK‑2 kidney proximal tubular cells were divided into 4 groups and treated with AngII (1x10‑6 M), HGF (8x10‑3 M), AngII plus HGF or control conditions, followed by an assessment of apoptosis induction and the expression levels of α‑smooth muscle actin (α‑SMA), which is a marker of TEMT. as well as the activation level of JAK2, phosphorylated (p)‑JAK2, STAT3 and p‑STAT3 signaling pathways. In HK‑2 cells, α‑SMA mRNA and protein expression levels increased following treatment with AngII, however, decreased expression was observed following exposure to HGF. HGF counteracted the AngII‑induced increase in the expression of α‑SMA in HK‑2 cells. Similar expression profiles were observed for the phosphorylated forms of JAK2 and STAT3, indicating the possible involvement of this signaling pathway. The results demonstrated that treatment of cells with AngII was associated with the induction of apoptosis when compared with the control. By contrast, treatment with HGF attenuated AngII‑induced apoptosis. The results suggested that HGF may inhibit TEMT by inhibiting AngII through the JAK2/STAT3 signaling pathway in HK‑2 cells and HGF may prevent apoptosis induced by AngII. The present study provides a basis for understanding the mechanisms involved in the inhibition of TEMT by HGF, which requires further investigation.
Collapse
Affiliation(s)
- Hong-Yue Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Chen Zhang
- Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, Fujian 361024, P.R. China
| | - Qing-Fei Xiao
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Hai-Chuan Dou
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Yan Chen
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Chun-Mei Gu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| | - Ming-Ji Cui
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130031, P.R. China
| |
Collapse
|
4
|
O'Connell PJ, Zhang W, Menon MC, Yi Z, Schröppel B, Gallon L, Luan Y, Rosales IA, Ge Y, Losic B, Xi C, Woytovich C, Keung KL, Wei C, Greene I, Overbey J, Bagiella E, Najafian N, Samaniego M, Djamali A, Alexander SI, Nankivell BJ, Chapman JR, Smith RN, Colvin R, Murphy B. Biopsy transcriptome expression profiling to identify kidney transplants at risk of chronic injury: a multicentre, prospective study. Lancet 2016; 388:983-93. [PMID: 27452608 PMCID: PMC5014570 DOI: 10.1016/s0140-6736(16)30826-1] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chronic injury in kidney transplants remains a major cause of allograft loss. The aim of this study was to identify a gene set capable of predicting renal allografts at risk of progressive injury due to fibrosis. METHODS This Genomics of Chronic Allograft Rejection (GoCAR) study is a prospective, multicentre study. We prospectively collected biopsies from renal allograft recipients (n=204) with stable renal function 3 months after transplantation. We used microarray analysis to investigate gene expression in 159 of these tissue samples. We aimed to identify genes that correlated with the Chronic Allograft Damage Index (CADI) score at 12 months, but not fibrosis at the time of the biopsy. We applied a penalised regression model in combination with permutation-based approach to derive an optimal gene set to predict allograft fibrosis. The GoCAR study is registered with ClinicalTrials.gov, number NCT00611702. FINDINGS We identified a set of 13 genes that was independently predictive for the development of fibrosis at 1 year (ie, CADI-12 ≥2). The gene set had high predictive capacity (area under the curve [AUC] 0·967), which was superior to that of baseline clinical variables (AUC 0·706) and clinical and pathological variables (AUC 0·806). Furthermore routine pathological variables were unable to identify which histologically normal allografts would progress to fibrosis (AUC 0·754), whereas the predictive gene set accurately discriminated between transplants at high and low risk of progression (AUC 0·916). The 13 genes also accurately predicted early allograft loss (AUC 0·842 at 2 years and 0·844 at 3 years). We validated the predictive value of this gene set in an independent cohort from the GoCAR study (n=45, AUC 0·866) and two independent, publically available expression datasets (n=282, AUC 0·831 and n=24, AUC 0·972). INTERPRETATION Our results suggest that this set of 13 genes could be used to identify kidney transplant recipients at risk of allograft loss before the development of irreversible damage, thus allowing therapy to be modified to prevent progression to fibrosis. FUNDING National Institutes of Health.
Collapse
Affiliation(s)
- Philip J O'Connell
- Renal Unit, University of Sydney at Westmead Hospital, Sydney, NSW, Australia
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madhav C Menon
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengzi Yi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bernd Schröppel
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lorenzo Gallon
- Department of Medicine-Nephrology and Surgery-Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yi Luan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bojan Losic
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Caixia Xi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher Woytovich
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karen L Keung
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilana Greene
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Overbey
- Department of Health Evidence and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emilia Bagiella
- Department of Health Evidence and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nader Najafian
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Milagros Samaniego
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Arjang Djamali
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Stephen I Alexander
- Discipline of Child & Adolescent Health, The Children's Hospital at Westmead Clinical School, The University of Sydney, NSW, Australia
| | - Brian J Nankivell
- Renal Unit, University of Sydney at Westmead Hospital, Sydney, NSW, Australia
| | - Jeremy R Chapman
- Renal Unit, University of Sydney at Westmead Hospital, Sydney, NSW, Australia
| | - Rex Neal Smith
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert Colvin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Barbara Murphy
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
5
|
Boor P, Floege J. Renal allograft fibrosis: biology and therapeutic targets. Am J Transplant 2015; 15:863-86. [PMID: 25691290 DOI: 10.1111/ajt.13180] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 11/30/2014] [Accepted: 12/19/2014] [Indexed: 01/25/2023]
Abstract
Renal tubulointerstitial fibrosis is the final common pathway of progressive renal diseases. In allografts, it is assessed with tubular atrophy as interstitial fibrosis/tubular atrophy (IF/TA). IF/TA occurs in about 40% of kidney allografts at 3-6 months after transplantation, increasing to 65% at 2 years. The origin of renal fibrosis in the allograft is complex and includes donor-related factors, in particular in case of expanded criteria donors, ischemia-reperfusion injury, immune-mediated damage, recurrence of underlying diseases, hypertensive damage, nephrotoxicity of immunosuppressants, recurrent graft infections, postrenal obstruction, etc. Based largely on studies in the non-transplant setting, there is a large body of literature on the role of different cell types, be it intrinsic to the kidney or bone marrow derived, in mediating renal fibrosis, and the number of mediator systems contributing to fibrotic changes is growing steadily. Here we review the most important cellular processes and mediators involved in the progress of renal fibrosis, with a focus on the allograft situation, and discuss some of the challenges in translating experimental insights into clinical trials, in particular fibrosis biomarkers or imaging modalities.
Collapse
Affiliation(s)
- P Boor
- Division of Nephrology and Clinical Immunology, RWTH University of Aachen, Aachen, Germany; Department of Pathology, RWTH University of Aachen, Aachen, Germany; Institute of Molecular Biomedicine, Bratislava, Slovakia
| | | |
Collapse
|
6
|
HGF-Met Pathway in Regeneration and Drug Discovery. Biomedicines 2014; 2:275-300. [PMID: 28548072 PMCID: PMC5344275 DOI: 10.3390/biomedicines2040275] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/15/2014] [Accepted: 10/13/2014] [Indexed: 12/26/2022] Open
Abstract
Hepatocyte growth factor (HGF) is composed of an α-chain and a β-chain, and these chains contain four kringle domains and a serine protease-like structure, respectively. Activation of the HGF–Met pathway evokes dynamic biological responses that support morphogenesis (e.g., epithelial tubulogenesis), regeneration, and the survival of cells and tissues. Characterizations of conditional Met knockout mice have indicated that the HGF–Met pathway plays important roles in regeneration, protection, and homeostasis in various cells and tissues, which includes hepatocytes, renal tubular cells, and neurons. Preclinical studies designed to address the therapeutic significance of HGF have been performed on injury/disease models, including acute tissue injury, chronic fibrosis, and cardiovascular and neurodegenerative diseases. The promotion of cell growth, survival, migration, and morphogenesis that is associated with extracellular matrix proteolysis are the biological activities that underlie the therapeutic actions of HGF. Recombinant HGF protein and the expression vectors for HGF are biological drug candidates for the treatment of patients with diseases and injuries that are associated with impaired tissue function. The intravenous/systemic administration of recombinant HGF protein has been well tolerated in phase I/II clinical trials. The phase-I and phase-I/II clinical trials of the intrathecal administration of HGF protein for the treatment of patients with amyotrophic lateral sclerosis and spinal cord injury, respectively, are ongoing.
Collapse
|
7
|
Kellenberger T, Marcussen N, Nyengaard JR, Wogensen L, Jespersen B. Expression of hypoxia-inducible factor-1α and hepatocyte growth factor in development of fibrosis in the transplanted kidney. Transpl Int 2014; 28:180-90. [DOI: 10.1111/tri.12475] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/21/2014] [Accepted: 10/02/2014] [Indexed: 01/30/2023]
Affiliation(s)
- Terese Kellenberger
- Research Laboratory for Biochemical Pathology; Department of Clinical Medicine; Aarhus University; Aarhus C Denmark
| | - Niels Marcussen
- Department of Pathology; Odense University Hospital; Odense C Denmark
| | - Jens R. Nyengaard
- Stereology and Electron Microscopy Laboratory; Centre for Stochastic Geometry and Advanced Bioimaging; Department of Clinical Medicine; Aarhus University; Aarhus C Denmark
| | - Lise Wogensen
- Research Laboratory for Biochemical Pathology; Department of Clinical Medicine; Aarhus University; Aarhus C Denmark
| | - Bente Jespersen
- Department of Renal Medicine; Aarhus University Hospital; Skejby Denmark
| |
Collapse
|
8
|
Shrestha BM, Haylor J. Biological pathways and potential targets for prevention and therapy of chronic allograft nephropathy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:482438. [PMID: 24971332 PMCID: PMC4058292 DOI: 10.1155/2014/482438] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/04/2014] [Indexed: 02/08/2023]
Abstract
Renal transplantation (RT) is the best option for patients with end-stage renal disease, but the half-life is limited to a decade due to progressive deterioration of renal function and transplant failure from chronic allograft nephropathy (CAN), which is the leading cause of transplant loss. Extensive research has been done to understand the pathogenesis, the biological pathways of fibrogenesis, and potential therapeutic targets for the prevention and treatment of CAN. Despite the advancements in the immunosuppressive agents and patient care, CAN continues to remain an unresolved problem in renal transplantation. The aim of this paper is to undertake a comprehensive review of the literature on the pathogenesis, biological pathways of RT fibrogenesis, and potential therapeutic targets for the prevention and therapy of CAN.
Collapse
Affiliation(s)
- Badri Man Shrestha
- Division of Renal Transplantation, Sheffield Kidney Institute, Northern General Hospital, Herries Road, Sheffield S5 7AU, UK
| | - John Haylor
- Division of Renal Transplantation, Sheffield Kidney Institute, Northern General Hospital, Herries Road, Sheffield S5 7AU, UK
| |
Collapse
|
9
|
Chang JW, Tsai HL, Chen CW, Yang HW, Yang AH, Yang LY, Wang PS, Ng YY, Lin TL, Lee OK. Conditioned mesenchymal stem cells attenuate progression of chronic kidney disease through inhibition of epithelial-to-mesenchymal transition and immune modulation. J Cell Mol Med 2014; 16:2935-49. [PMID: 22862802 PMCID: PMC4393722 DOI: 10.1111/j.1582-4934.2012.01610.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 07/24/2012] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been shown to improve the outcome of acute renal injury models; but whether MSCs can delay renal failure in chronic kidney disease (CKD) remains unclear. In the present study, the were cultured in media containing various concentrations of basic fibroblast growth factor, epidermal growth factor and ascorbic acid 2-phosphate to investigate whether hepatocyte growth factor (HGF) secretion could be increased by the stimulation of these growth factors. Then, TGF-β1-treated renal interstitial fibroblast (NRK-49F), renal proximal tubular cells (NRK-52E) and podocytes were co-cultured with conditioned MSCs in the absence or presence of ascorbic acid 2-phosphate to quantify the protective effects of conditioned MSCs on renal cells. Moreover, male Sprague-Dawley rats were treated with 1 × 106 conditioned MSCs immediately after 5/6 nephrectomy and every other week through the tail vein for 14 weeks. It was found that basic fibroblast growth factor, epidermal growth factor and ascorbic acid 2-phosphate promoted HGF secretion in MSCs. Besides, conditioned MSCs were found to be protective against TGF-β1 induced epithelial-to-mesenchymal transition of NRK-52E and activation of NRK-49F cells. Furthermore, conditioned MSCs protected podocytes from TGF-β1-induced loss of synaptopodin, fibronectin induction, cell death and apoptosis. Rats transplanted with conditioned human MSCs had a significantly increase in creatinine clearance rate, decrease in glomerulosclerosis, interstitial fibrosis and increase in CD4+CD25+Foxp3+ regulatory T cells counts in splenocytes. Together, our studies indicated that conditioned MSCs preserve renal function by their anti-fibrotic and anti-inflammatory effects. Transplantation of conditioned MSCs may be useful in treating CKD.
Collapse
Affiliation(s)
- Jei-Wen Chang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wang HY, Yang LZ, Gu CM, Chen Y, Zhao Y, Zhao D, Li TS, Cui MJ. Pathological changes, TGF-β1 expression, and the effects of hepatocyte growth factor in 5/6 nephrectomized rats. Ren Fail 2013; 36:393-9. [PMID: 24345300 DOI: 10.3109/0886022x.2013.867797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Several studies have shown that hepatocyte growth factor (HGF) ameliorates chronic renal failure, but its mechanism of action is unclear. This study was designed to test the delivery of HGF in the PCI-neo vector, using the 5/6 nephrectomized rat as a model for chronic renal failure, and to confirm that this protective function is associated with decreased protein expression of transforming growth factor-beta1 (TGF-β1). Rats were randomly divided into the following groups: Control (untreated), PCI-neo (vector control), 5/6 nephrectomy, and PCI-neo-HGF. Rats were sacrificed at both the fifth and ninth week after 5/6 nephrectomy. Kidney specimens were used for pathological examination (hematoxylin-eosin staining), and detection of TGF-β1 protein (Western blot and immunohistochemistry) expression. Blood urea nitrogen, serum creatinine, and 24-h urinary protein excretion (UPE) were increased, renal interstitium was seriously injured, and TGF-β1 protein expression was elevated in 5/6 nephrectomized rats compared to control rats at either time point. Red blood cell and hemoglobin levels decreased in the ninth week after 5/6 nephrectomy. PCI-neo-HGF expression ameliorated the aforementioned changes and decreased TGF-β1 expression, not only in the fifth week, but also in the ninth week after surgery. The process of renal injury in the 5/6 nephrectomized rat was consistent with that of chronic renal failure. The increase in TGF-β1 expression was maintained after 5/6 nephrectomy. HGF relieved chronic renal failure, this protection was associated with down-regulation of TGF-β1 protein expression, and the protective effects were long-term and stable after 5/6 nephrectomy.
Collapse
Affiliation(s)
- Hong-Yue Wang
- Department of Nephrology, First Hospital of Jilin University , Changchun , China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Xu D, Zhang T, Chen X, Zhou Q, Liu C, Deng Z, Zhang L, Ying C, Zhang W, Gu M. Reduction of osteopontin in vivo inhibits tubular epithelial to mesenchymal transition in rats with chronic allograft nephropathy. Transplant Proc 2013; 45:659-65. [PMID: 23498805 DOI: 10.1016/j.transproceed.2012.02.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 02/13/2012] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Chronic allograft nephropathy (CAN) is an important etiological factor causing graft loss. However, the mechanism of CAN is unclear. Osteopontin (OPN), a proinflammatory and profibrosis molecule, plays a key role in late stages of renal diseases. We investigated the potential role of OPN in the pathogenesis of CAN. METHODS Using a F344 to Lewis rat CAN model, we injected short hairpin RNA (shRNA) constructs targeting OPN or negative control plasmids through the renal vein following electroporation. At 12 weeks after the transplantation, we determined interstitial fibrosis (IF) and tubular atrophy (TA) of the tubular epithelial cells (TECs). OPN expression was examined using Western blots and immunohistochemistry (IHC). Molecules involved in epithelial to mesenchymal transition (EMT) of TECs were examined using IHC and Western blots. RESULTS OPN expression in kidney grafts was decreased by the RNA interference (RNAi) group. Histology observations showed IF and TA to be mild with stable renal function in the RNAi-treated group. EMT of TECs was significantly lessened after reducing OPN. CONCLUSION Reduction of OPN in vivo inhibited progression of CAN. OPN may be of therapeutic value in transplantation settings.
Collapse
Affiliation(s)
- D Xu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Dosanjh A, Robison E, Mondala T, Head SR, Salomon DR, Kurian SM. Genomic meta-analysis of growth factor and integrin pathways in chronic kidney transplant injury. BMC Genomics 2013; 14:275. [PMID: 23617750 PMCID: PMC3644490 DOI: 10.1186/1471-2164-14-275] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 04/18/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Chronic Allograft Nephropathy (CAN) is a clinical entity of progressive kidney transplant injury. The defining histology is tubular atrophy with interstitial fibrosis (IFTA). Using a meta-analysis of microarrays from 84 kidney transplant biopsies, we revealed growth factor and integrin adhesion molecule pathways differentially expressed and correlated with histological progression. A bioinformatics approach mining independent datasets leverages new and existing data to identify correlative changes in integrin and growth factor signaling pathways. RESULTS Analysis of CAN/IFTA Banff grades showed that hepatocyte growth factor (HGF), and epidermal growth factor (EGF) pathways are significantly differentially expressed in all classes of CAN/IFTA. MAPK-dependent pathways were also significant. However, the TGFβ pathways, albeit present, failed to differentiate CAN/IFTA progression. The integrin subunits β8, αv, αμ and β5 are differentially expressed, but β1, β6 and α6 specifically correlate with progression of chronic injury. Results were validated using our published proteomic profiling of CAN/IFTA. CONCLUSIONS CAN/IFTA with chronic kidney injury is characterized by expression of distinct growth factors and specific integrin adhesion molecules as well as their canonical signaling pathways. Drug target mapping suggests several novel candidates for the next generation of therapeutics to prevent or treat progressive transplant dysfunction with interstitial fibrosis.
Collapse
Affiliation(s)
- Amrita Dosanjh
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
13
|
Li Z, Chen J, Li L, Ran JH, Li XH, Liu ZH, Liu GJ, Gao YC, Zhang XL, Sun HD. Human hepatocyte growth factor (hHGF)-modified hepatic oval cells improve liver transplant survival. PLoS One 2012; 7:e44805. [PMID: 23028627 PMCID: PMC3445612 DOI: 10.1371/journal.pone.0044805] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 08/14/2012] [Indexed: 01/14/2023] Open
Abstract
Despite progress in the field of immunosuppression, acute rejection is still a common postoperative complication following liver transplantation. This study aims to investigate the capacity of the human hepatocyte growth factor (hHGF) in modifying hepatic oval cells (HOCs) administered simultaneously with orthotopic liver transplantation as a means of improving graft survival. HOCs were activated and isolated using a modified 2-acetylaminofluorene/partial hepatectomy (2-AAF/PH) model in male Lewis rats. A HOC line stably expressing the HGF gene was established following stable transfection of the pBLAST2-hHGF plasmid. Our results demonstrated that hHGF-modified HOCs could efficiently differentiate into hepatocytes and bile duct epithelial cells in vitro. Administration of HOCs at the time of liver transplantation induced a wider distribution of SRY-positive donor cells in liver tissues. Administration of hHGF-HOC at the time of transplantation remarkably prolonged the median survival time and improved liver function for recipients compared to these parameters in the other treatment groups (P<0.05). Moreover, hHGF-HOC administration at the time of liver transplantation significantly suppressed elevation of interleukin-2 (IL-2), tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) levels while increasing the production of IL-10 and TGF-β1 (P<0.05). HOC or hHGF-HOC administration promoted cell proliferation, reduced cell apoptosis, and decreased liver allograft rejection rates. Furthermore, hHGF-modified HOCs more efficiently reduced acute allograft rejection (P<0.05 versus HOC transplantation only). Our results indicate that the combination of hHGF-modified HOCs with liver transplantation decreased host anti-graft immune responses resulting in a reduction of allograft rejection rates and prolonging graft survival in recipient rats. This suggests that HOC-based cell transplantation therapies can be developed as a means of treating severe liver injuries.
Collapse
Affiliation(s)
- Zhu Li
- Department of Hepatobiliary Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
He D, Lee L, Yang J, Wang X. Preventive effects and mechanisms of rhein on renal interstitial fibrosis in obstructive nephropathy. Biol Pharm Bull 2012; 34:1219-26. [PMID: 21804209 DOI: 10.1248/bpb.34.1219] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Renal interstitial fibrosis is a common outcome of a variety of chronic renal diseases. Here we evaluated the therapeutic efficacy of rhein on renal interstitial fibrosis induced by unilateral ureteral obstruction (UUO) and investigated the potential mechanisms. Mice underwent UUO, followed by orally administrated rhein (150 mg/kg/d) or control vehicle. Renal interstitial injury and the degree of fibrosis were evaluated by pathological staining and Western blot. The possible mechanisms were studied by Western blot, indirect immune-fluorescence and enzyme-linked immunosorbent assay. Our results showed that rhein therapy markedly ameliorated renal interstitial fibrotic lesions, reduced α-smooth muscle actin (α-SMA) expression, attenuated deposition of fibronectin (FN). Rhein also suppressed transforming growth factor-β1 (TGF-β1) and its type I receptor expression in obstructed kidneys. In vitro, rhein abolished the α-SMA and fibronectin expression of rat kidney interstitial fibroblasts cells (NRK-49F) induced by TGF-β1. These observations strongly suggest that rhein is a potent inhibitor of renal interstitial fibrosis, and its therapeutic mechanism is, at least in part, blocking interstitial fibroblasts cells activation.
Collapse
Affiliation(s)
- Dongyuan He
- First Clinical Medical College of Nanjing Medical University, Nanjing 210029, P. R. China
| | | | | | | |
Collapse
|
15
|
Agustian PA, Schiffer M, Gwinner W, Schäfer I, Theophile K, Modde F, Bockmeyer CL, Traeder J, Lehmann U, Grosshennig A, Kreipe HH, Bröcker V, Becker JU. Diminished met signaling in podocytes contributes to the development of podocytopenia in transplant glomerulopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2007-19. [PMID: 21514418 DOI: 10.1016/j.ajpath.2011.01.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 12/19/2010] [Accepted: 01/21/2011] [Indexed: 01/24/2023]
Abstract
Transplant glomerulopathy (TxG) can show secondary focal and segmental glomerulosclerosis (FSGS). FSGS in native kidneys is caused by podocytopenia. This study examines podocytopenia and the role of decreased paracrine Met activation on podocytes by decreased glomerular hepatocyte growth factor (HGF) levels in the development of podocytopenia in TxG. Podocytes were counted in 10 zero-hour biopsies and 10 specimens each with and without TxG. HGF/Met was examined with immunostains and quantitative RT-PCR in a set of three consecutive biopsies from 10 patients with TxG, including the diagnostic biopsy (DiagnBx) and the two previous biopsies (1stPrevBx and 2ndPrevBx). Antiapoptotic effects of HGF on podocytes were examined in vitro. Mean podocyte numbers per glomerulus were lower and glomerular volume higher in TxG. Fewer of the two preceding biopsies of the patients than of the controls contained phospho-Met(Tyr1349)-positive podocytes (2 of 8 versus 7 of 7, P = 0.0070; 4 of 9 versus 9 of 9, P = 0.0294). Glomerular HGF mRNA levels were lower in the 1stPrevBx of the patients (0.049 ± 0.083 versus 0.284 ± 0.331; P = 0.0155). In vitro, HGF stimulation of podocytes resulted in antiapoptotic phosphorylation of AKT and extracellular signal-regulated kinase (ERK) and induction of X-linked inhibitor of apoptosis protein (XIAP). Decreased antiapoptotic Met signaling in podocytes, probably due to decreased HGF secretion by glomerular epithelial cells, could contribute to podocyte loss and FSGS in TxG.
Collapse
Affiliation(s)
- Putri A Agustian
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kato T, Mizuno S, Nakamura T. Preservations of nephrin and synaptopodin by recombinant hepatocyte growth factor in podocytes for the attenuations of foot process injury and albuminuria in nephritic mice. Nephrology (Carlton) 2011; 16:310-8. [PMID: 21342325 DOI: 10.1111/j.1440-1797.2010.01392.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIM Podocytes provide a slit diaphragm to inhibit proteinuria, and nephrin between podocytes functions as a barrier during glomerular filtration. Hepatocyte growth factor (HGF) can improve proteinuria in rodents with various renal injuries, but little is known about the role of HGF in podocyte-based events during glomerulonephritis. In the present study, we examined whether and how nephrin expression is sustained by podocytes during the HGF-mediated attenuation of albuminuria. METHODS Lipopolysaccharide (LPS)-treated mice were used as an animal model of albuminuria. We evaluated the effect of HGF on slit proteins using immunohistochemistry, western blotting and real-time polymerase chain reaction. RESULTS Albuminuria occurred 36 h after LPS treatment in mice. This albuminuria did not involve podocyte loss, but was associated with a decrease in nephrin and its key anchor, synaptopodin. In these processes, c-Met tyrosine phosphorylation, which represented HGF signal activation, occurred in glomerular cells including podocytes. When recombinant HGF was administrated to nephritic mice, c-Met tyrosine phosphorylation became evident in podocytes. The enhancement of the HGF-c-Met signal was associated with increases in nephrin and synaptopodin. An electron microscopic examination revealed that LPS induced the foot process effacement of podocytes, while HGF injections suppressed the foot process injury. Overall, albuminuria was attenuated in the LPS-treated mice after HGF administration. CONCLUSION HGF protects podocytes from a loss of nephrin, at least in part, through maintaining synaptopodin. As a result, HGF was shown to sustain foot process structure, and albuminuria was attenuated under inflammation.
Collapse
Affiliation(s)
- Takashi Kato
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | |
Collapse
|
17
|
Azuma H, Isaka Y, Nomi H, Inamoto T, Li XK, Hounig T, Takabatake Y, Ichimaru N, Ibuki N, Matsumoto K, Ubai T, Katsuoka Y, Takahara S. Induction of donor-specific tolerance using superagonistic CD28 antibody in rat renal allografts: regulatory T-cell expansion before engraftment may be important. Transplantation 2011; 90:1328-35. [PMID: 21085062 DOI: 10.1097/tp.0b013e3182007b59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND We hypothesized that a superagonistic monoclonal antibody specific for CD28 (CD28SA), which expands regulatory T cells (Tregs) in vivo, would prevent acute rejection and prolong the survival of renal allograft. METHODS We examined whether CD28SA treatment induce donor-specific tolerance using our established rat renal allograft model (Wistar-Lewis). RESULTS All control rats died within 13 days because of severe azotemia with marked destruction of graft tissue. In contrast, recipients treated with a triple injection of CD28SA (days -3, 0, and 3) showed good preservation of graft histology and function, with considerable infiltration of Tregs into the allografts; 92% of recipients survived for more than 100 days, and 77% survived by the day of harvest at 180 days. These long-surviving recipients received secondary heterotopic bicardiac allografts from both donor-matched Wistar and third-party Brown Norway rats simultaneously 120 days after kidney transplantation, and seven of eight (87.5%) rats exhibited donor-specific tolerance, accepting the Wistar heart, but acutely rejecting the Brown Norway heart. Interestingly, a single injection of CD28SA 3 days before (day -3), but not 3 days after (day 3), transplantation also induced donor-specific tolerance in some recipients. We then performed adoptive transfer of nonspecific CD4+CD25+ Tregs, purified from CD28SA-treated Lewis rats, with simultaneous injection of hepatocyte growth factor (500 μg/kg/day, intravenously). The treatment induced significant prolongation of graft survival (P<0.0001 vs. control group), and five of eight (62.5%) recipients survived until the day of harvest at 180 days with successful induction of donor-specific tolerance. CONCLUSIONS We have established a novel therapeutic approach for inducing donor-specific tolerance in rats with renal allografts.
Collapse
Affiliation(s)
- Haruhito Azuma
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Furuichi K, Wada T, Kaneko S. Cytokines and chemokines as therapeutic targets for ischemic kidney injury. Inflamm Regen 2011. [DOI: 10.2492/inflammregen.31.365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
19
|
Dai C, Saleem MA, Holzman LB, Mathieson P, Liu Y. Hepatocyte growth factor signaling ameliorates podocyte injury and proteinuria. Kidney Int 2010; 77:962-973. [PMID: 20375988 PMCID: PMC3071594 DOI: 10.1038/ki.2010.40] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hepatocyte growth factor (HGF) is a potent antifibrotic protein that inhibits kidney fibrosis through several mechanisms. To study its role in podocyte homeostasis, injury, and repair in vivo, we generated conditional knockout mice in which the HGF receptor, c-met, was specifically deleted in podocytes using the Cre-LoxP system. Mice with podocyte-specific ablation of c-met (podo-met(-/-)) developed normally. No albuminuria or overt pathologic lesions were detected up to 6 months of age, suggesting that HGF signaling is dispensable for podocyte maturation, survival, and function under normal physiologic conditions. However, after adriamycin treatment, podo-met(-/-) mice developed more severe podocyte injury and albuminuria than their control littermates. Ablation of c-met also resulted in more profound suppression of Wilms tumor 1 (WT1) and nephrin expression, and podocyte apoptosis after injury. When HGF was expressed ectopically in vivo, it ameliorated adriamycin-induced albuminuria, preserved WT1 and nephrin expression, and inhibited podocyte apoptosis. However, exogenous HGF failed to significantly reduce albuminuria in podo-met(-/-) mice, suggesting that podocyte-specific c-met activation by HGF confers renal protection. In vitro, HGF was able to preserve WT1 and nephrin expression in cultured podocytes after adriamycin treatment. HGF also protected podocytes from apoptosis induced by a lethal dose of adriamycin primarily through a phosphoinositide 3-kinase (PI3K)/Akt-dependent pathway. Collectively, these results indicate that HGF/c-met signaling has an important role in protecting podocytes from injury, thereby reducing proteinuria.
Collapse
Affiliation(s)
- Chunsun Dai
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Moin A. Saleem
- Children’s Renal Unit, Bristol Royal Hospital for Children, Bristol, UK
| | - Lawrence B. Holzman
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Abstract
Kidney allograft fibrosis results from a reactive process mediated by humoral and cellular events and the activation of transforming growth factor beta1. It is a process that involves both parenchymal and graft infiltrating cells and can lead to organ failure if injury persists or if the response to injury is excessive. In this review, we will address the role of preventive and therapeutic strategies that target kidney allograft fibrogenesis. We conclude that in addition to preventive strategies, therapies based on bone morphogenetic protein 7, hepatocyte growth factor, connective tissue growth factor, and pirfenidone have shown promising results in preclinical studies. Clinical trials are needed to examine the effect of these therapies on long-term outcomes.
Collapse
|
21
|
Nakamura T, Mizuno S. The discovery of hepatocyte growth factor (HGF) and its significance for cell biology, life sciences and clinical medicine. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2010; 86:588-610. [PMID: 20551596 PMCID: PMC3081175 DOI: 10.2183/pjab.86.588] [Citation(s) in RCA: 374] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
It has been more than 25 years since HGF was discovered as a mitogen of hepatocytes. HGF is produced by stromal cells, and stimulates epithelial cell proliferation, motility, morphogenesis and angiogenesis in various organs via tyrosine phosphorylation of its receptor, c-Met. In fetal stages, HGF-neutralization, or c-Met gene destruction, leads to hypoplasia of many organs, indicating that HGF signals are essential for organ development. Endogenous HGF is required for self-repair of injured livers, kidneys, lungs and so on. In addition, HGF exerts protective effects on epithelial and non-epithelial organs (including the heart and brain) via anti-apoptotic and anti-inflammatory signals. During organ diseases, plasma HGF levels significantly increased, while anti-HGF antibody infusion accelerated tissue destruction in rodents. Thus, endogenous HGF is required for minimization of diseases, while insufficient production of HGF leads to organ failure. This is the reason why HGF supplementation produces therapeutic outcomes under pathological conditions. Moreover, emerging studies delineated key roles of HGF during tumor metastasis, while HGF-antagonism leads to anti-tumor outcomes. Taken together, HGF-based molecules, including HGF-variants, HGF-fragments and c-Met-binders are available as regenerative or anti-tumor drugs. Molecular analysis of the HGF-c-Met system could provide bridges between basic biology and clinical medicine.
Collapse
Affiliation(s)
- Toshikazu Nakamura
- Kringle Pharma Joint Research Division for Regenerative Drug Discovery, Center for Advanced Science and Innovation, Osaka University, Osaka, Japan.
| | | |
Collapse
|
22
|
Nagano T, Mori-Kudo I, Kawamura T, Taiji M, Noguchi H. Pre‐ or Post‐treatment with Hepatocyte Growth Factor Prevents Glycerol‐Induced Acute Renal Failure. Ren Fail 2009; 26:5-11. [PMID: 15083915 DOI: 10.1081/jdi-120028537] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hepatocyte growth factor (HGF) is known to have beneficial effects against damage in various organs, including liver, kidney and lung, in disease models. Previously, we reported that repeated administration of HGF ameliorates renal dysfunction and histological alteration of glycerol-injected rats, an animal model for severe acute renal failure (ARF). In the present study, we investigated in more detail the efficacy of pre- and post-treatment of HGF in this model. ARF was induced by intramuscular injection of glycerol into the hind limbs of male Wistar rats. The efficacy of pre-treatment was studied by intravenous injection of HGF (1 mg/kg) or vehicle 1 and 18 hours prior to glycerol injection. Pre-treatment of HGF dramatically protected glycerol-induced ARF rats against death, and prevented deterioration of biochemical parameters for renal function. We also analyzed expression of heme oxygenase-1 (HO-1), a cytoprotective protein, in kidney of HGF-injected rats. Intravenous administration of HGF enhanced renal expression of HO-1 mRNA from 1 to 3 hours after injection. Next, as a post-treatment study, HGF (1 mg/kg/3 hours) with dopamine was infused into glycerol-induced ARF rats 7 hours after glycerol injection. Intravenous infusion of HGF after ARF onset also ameliorated renal biochemical parameters. These results indicate that pre-treatment of HGF can improve ARF, and induction of HO-1 expression in kidney may be a cause of the protective effect. In addition, post-treatment of HGF with dopamine was also effective against the establishment of ARF.
Collapse
Affiliation(s)
- Tomokazu Nagano
- Discovery Research Laboratories I, Sumitomo Pharmaceuticals Research Division, Konohana-ku, Osaka, Japan
| | | | | | | | | |
Collapse
|
23
|
Rampino T, Gregorini M, Dal Canton A. Scatter Factors in renal disease: Dr. Jeckyll and Mr. Hyde? Cytokine Growth Factor Rev 2009; 20:77-85. [PMID: 19201250 DOI: 10.1016/j.cytogfr.2009.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The Scatter Factors are two homologous proteins, named Scatter Factor/Hepatocyte Growth Factor and Macrophage Stimulating Protein. Their receptors are the products of two oncogenes, Met and Ron, respectively. The Scatter Factors induce movement, stimulate proliferation, regulate apoptosis and are morphogenic, i.e. operate an integrated program that seems tailored to drive organ development and to regenerate injured tissues. On the other hand, Scatter Factors may be responsible for pathologic tissue remodeling, infiltration of inflammatory cells, and tumor growth and diffusion. The review describes the involvement of Scatter Factors in renal disease, including acute renal failure, glomerulonephritis, chronic fibrosing nephropathies, dialysis, renal transplantation and renal tumors, and discusses the double-faced role of Scatter Factors, that play either a protective or a pathogenic role.
Collapse
Affiliation(s)
- Teresa Rampino
- Foundation IRCCS San Matteo Hospital and University of Pavia, Italy
| | | | | |
Collapse
|
24
|
Beckmann N, Cannet C, Babin AL, Blé F, Zurbruegg S, Kneuer R, Dousset V. In vivo
visualization of macrophage infiltration and activity in inflammation using magnetic resonance imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2009; 1:272-98. [DOI: 10.1002/wnan.16] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Nicolau Beckmann
- Global Imaging Group, Novartis Institutes for BioMedical Research, CH‐4056 Basel, Switzerland
| | - Catherine Cannet
- Global Imaging Group, Novartis Institutes for BioMedical Research, CH‐4056 Basel, Switzerland
| | - Anna Louise Babin
- Global Imaging Group, Novartis Institutes for BioMedical Research, CH‐4056 Basel, Switzerland
- Respiratory Diseases Department, Novartis Institutes for BioMedical Research, CH‐4056 Basel, Switzerland
- Sackler Institute of Pulmonary Pharmacology, King's College, London SE1 1UL, UK
| | - François‐Xavier Blé
- Respiratory Diseases Department, Novartis Institutes for BioMedical Research, CH‐4056 Basel, Switzerland
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Canada M5T 3H7
| | - Stefan Zurbruegg
- Global Imaging Group, Novartis Institutes for BioMedical Research, CH‐4056 Basel, Switzerland
| | - Rainer Kneuer
- Global Imaging Group, Novartis Institutes for BioMedical Research, CH‐4056 Basel, Switzerland
| | - Vincent Dousset
- University Victor Segalen Bordeaux 2, EA 2966 Neurobiology of Myelin Disease Laboratory, CHU de Bordeaux, F‐33076 Bordeaux, France
| |
Collapse
|
25
|
Shukla MN, Rose JL, Ray R, Lathrop KL, Ray A, Ray P. Hepatocyte growth factor inhibits epithelial to myofibroblast transition in lung cells via Smad7. Am J Respir Cell Mol Biol 2008; 40:643-53. [PMID: 18988920 DOI: 10.1165/rcmb.2008-0217oc] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Idiopathic pulmonary fibrosis is a lethal parenchymal lung disease characterized by denudation of the lung epithelium, fibroblast proliferation, and collagen deposition. Cellular changes underlying disease progression involve injury to alveolar epithelial cells, epithelial to mesenchymal transition, proliferation of alpha-smooth muscle actin (alpha-SMA)-expressing myofibroblasts and of fibroblasts resulting in enhanced deposition of extracellular matrix proteins. Hepatocyte growth factor (HGF) inhibits progression of bleomycin-induced pulmonary fibrosis in mice. The mechanism underlying the inhibitory effect of HGF was investigated in an in vitro model. We show that HGF markedly antagonizes basal and transforming growth factor (TGF)-beta-induced expression of myofibroblast markers such as alpha-SMA, collagen type 1, and fibronectin in rat alveolar epithelial cells. HGF also inhibited TGF-beta-induced alpha-SMA expression in primary murine alveolar epithelial cells. Since TGF-beta is known to regulate alpha-SMA expression, the effect of HGF on components of TGF-beta signaling was investigated. HGF induced expression of Smad7, an inhibitor of TGF-beta signaling, in a mitogen-activated protein kinase-dependent manner. HGF also induced the nuclear export of Smad7 and Smad ubiquitin regulatory factor 1 (Smurf1) to the cytoplasm. HGF-dependent decrease in alpha-SMA was abolished with specific siRNAs targeted to Smad7. Thus, induction of Smad7 by HGF serves to limit acquisition of the myofibroblast phenotype in alveolar epithelial cells.
Collapse
Affiliation(s)
- Manasi N Shukla
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | |
Collapse
|
26
|
Giannopoulou M, Dai C, Tan X, Wen X, Michalopoulos GK, Liu Y. Hepatocyte growth factor exerts its anti-inflammatory action by disrupting nuclear factor-kappaB signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:30-41. [PMID: 18502824 PMCID: PMC2438283 DOI: 10.2353/ajpath.2008.070583] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/31/2008] [Indexed: 12/21/2022]
Abstract
Renal inflammation, characterized by the influx of inflammatory cells, is believed to play a critical role in the initiation and progression of a wide range of chronic kidney diseases. Here, we show that hepatocyte growth factor (HGF) inhibited renal inflammation and proinflammatory chemokine expression by disrupting nuclear factor (NF)-kappaB signaling. In vivo, HGF gene delivery inhibited interstitial infiltration of inflammatory T cells and macrophages, and suppressed expression of both RANTES (regulated on activation, normal T cell expressed and secreted) and monocyte chemoattractant protein-1 in a mouse model of obstructive nephropathy. In vitro, HGF abolished RANTES induction in human kidney epithelial cells, which was dependent on NF-kappaB signaling. HGF did not significantly affect the phosphorylation or degradation of IkappaBalpha; it also did not influence the phosphorylation or nuclear translocation of p65 NF-kappaB. However, HGF prevented p65 NF-kappaB binding to its cognate cis-acting element in the RANTES promoter. HGF action was dependent on the activation of the phosphoinositide 3-kinase/Akt pathway, which led to the phosphorylation and inactivation of glycogen synthase kinase (GSK)-3beta. Suppression of GSK-3beta activity mimicked HGF and abolished RANTES expression, whereas ectopic expression of GSK-3beta restored RANTES induction. HGF also induced renal GSK-3beta phosphorylation and inactivation after obstructive injury in vivo. These observations suggest that HGF is a potent anti-inflammatory cytokine that inhibits renal inflammation by disrupting NF-kappaB signaling and may be a promising therapeutic agent for progressive renal diseases.
Collapse
Affiliation(s)
- Myrto Giannopoulou
- Department of Pathology, University of PittsburghSchool of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | | | |
Collapse
|
27
|
Jevnikar AM, Mannon RB. Late kidney allograft loss: what we know about it, and what we can do about it. Clin J Am Soc Nephrol 2008; 3 Suppl 2:S56-67. [PMID: 18309004 DOI: 10.2215/cjn.03040707] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite dramatic improvements in immunosuppression, late graft loss after kidney transplantation remains a common and difficult problem. Histologic evaluation may reveal changes related to BK polyomavirus infection, hypertension, or calcineurin inhibitor toxicity, which can help to guide therapy. The designation chronic allograft nephropathy should thus be reserved for biopsies with tubular atrophy and interstitial fibrosis without an apparent cause. Although the cause clearly includes both antigen-dependent and antigen-independent events, the approach remains largely to exclude immune mechanisms. Although this review discusses the potential contribution of antibody to chronic injury, it focuses on the basic elements of kidney injury, the role of parenchymal cells in promoting injury, and the proliferative and inflammatory responses that accompanying injury. Strategies to manage these recipients include close attention to accompanying hypertension, diabetes, and hyperlipidemia, as well as consideration for altering immunosuppression; however, therapies that limit epithelial-to-mesenchymal transition or directly block fibrosis pathways may reduce chronic allograft fibrosis and may prove to be useful. Understanding the basic pathogenesis sufficiently to allow early intervention may finally benefit patients who are at high risk for tubular atrophy and interstitial fibrosis and promote their long-term graft function.
Collapse
Affiliation(s)
- Anthony M Jevnikar
- Department of Medicine, Lawson Health Research Institute and Robarts Research Institute, London Health Sciences Centre, University of Western Ontario, London, Ontario, Canada
| | | |
Collapse
|
28
|
Mesenchymal stem cells modulate immune responses combined with cyclosporine in a rat renal transplantation model. Transplant Proc 2008; 39:3404-8. [PMID: 18089393 DOI: 10.1016/j.transproceed.2007.06.092] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2007] [Accepted: 06/21/2007] [Indexed: 12/26/2022]
Abstract
Mesenchymal stem cells (MSCs) are pluripotent progenitors for a variety of cell types. Their down-regulation of the immune response in vivo has been hypothesized, but not yet substantiated, by experimental evidence. We investigated graft function, histology, animal survival days reverse transcriptase-polymerase chain reactor (RT-PCR) analysis of interleukin (IL)-1, tumor necrosis factor (TNF)-alpha, and transforming growth factor (TGF)-beta1. The results demonstrated that MSCs down-regulated immune responses, reduced production of some inflammatory mediators, preserved graft function in the initial stage after transplantation, and prolonged animal survival. However, the effects were not as strong as those of cyclosporine (CsA) therapy. Moreover, MSCs combined with low-dose CsA protected graft function, but could not prolong animal survival compared with CsA monotherapy, indicating a potential interaction between MSC and CsA activities, possibly allowing reduced CsA doses.
Collapse
|
29
|
Mizuno S, Nakamura T. Hepatocyte growth factor: a regenerative drug for acute hepatitis and liver cirrhosis. Regen Med 2007; 2:161-70. [PMID: 17465748 DOI: 10.2217/17460751.2.2.161] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Liver cirrhosis is a major cause of morbidity worldwide and is characterized by the loss of hepatocytes with interstitial fibrosis. In this review, we discuss the potential uses of hepatocyte growth factor for treating hepatic diseases, focusing on the molecular mechanisms whereby hepatocyte growth factor reverses liver cirrhosis. Hepatic myofibroblasts play a central role in the development of liver cirrhosis, while myofibroblasts acquire c-Met. Using a rat model of liver cirrhosis, we recently delineated the direct effect of hepatocyte growth factor toward myofibroblasts: the induction of apoptotic cell death associated with matrix degradation, the inhibition of overproliferation and the suppression of transforming growth factor-beta1 production in myofibroblasts. Hepatocyte growth factor elicits mitogenic, anti-apoptotic and anti-inflammatory functions in hepatocytes, therefore contributing to reversing liver dysfunction. Considering the insufficient production of hepatocyte growth factor is responsible for the manifestation of chronic hepatitis, supplementation with or reinduction of hepatocyte growth factor represents a new strategy for attenuating intractable liver diseases.
Collapse
Affiliation(s)
- Shinya Mizuno
- Division of Molecular Regenerative Medicine, Department of Biochemistry and Molecular Biology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, Japan
| | | |
Collapse
|
30
|
Abstract
The increasing global prevalence of chronic kidney disease (CKD) and end-stage renal disease with the associated spiraling cost has profound public health and economic implications. This has made slowing the progression of CKD, a major health-care priority. CKD is invariably characterized by progressive kidney fibrosis and at present, treatment aiming to slow the progression of CKD is limited to aggressive blood pressure control, with few therapies targeting the fibrotic process itself. In this review, we explore the potential of experimental therapeutic strategies, based on preventing or reversing the pathophysiologic steps of kidney remodeling that lead to fibrosis.
Collapse
Affiliation(s)
- A Khwaja
- Department of Academic Nephrology, Sheffield Kidney Institute, Sheffield, UK.
| | | | | | | |
Collapse
|
31
|
Tan R, Zhang X, Yang J, Li Y, Liu Y. Molecular basis for the cell type specific induction of SnoN expression by hepatocyte growth factor. J Am Soc Nephrol 2007; 18:2340-2349. [PMID: 17625116 DOI: 10.1681/asn.2007010128] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hepatocyte growth factor (HGF) is a potent antifibrotic cytokine that antagonizes the TGF-beta1/Smad signaling in diverse types of kidney cells by different mechanisms. HGF is shown to induce Smad co-repressor Sloan-Kettering Institute proto-oncogene-related novel gene, non-Alu-containing (SnoN) expression in proximal tubular epithelial cells (HKC-8) but not in glomerular mesangial cells and interstitial fibroblasts. This study investigated the molecular mechanisms underlying the cell type-specific induction of SnoN by HGF. Treatment of HKC-8 cells with actinomycin D completely abolished HGF-mediated SnoN induction, suggesting its dependence on gene transcription. Although HGF activated several signal pathways in HKC-8 cells, blockade of extracellular signal-regulated kinase-1 and -2 (Erk-1/2) activation but not Akt and p38 mitogen-activated protein kinase abolished SnoN induction. HGF rapidly activated both upstream and downstream signaling of Erk-1/2, which led to the activation of the cAMP response element-binding protein (CREB). In the promoter region of human SnoN gene, two cAMP response elements were located in close proximity to Sp1 sites. Chromatin immunoprecipitation assay revealed that activated CREB and Sp1 bound to their cognate cis-acting elements in SnoN promoter in response to HGF stimulation. Ectopic expression of wild-type CREB promoted SnoN expression, whereas dominant negative mutant CREB abrogated SnoN induction by HGF. Likewise, chemical blockade of Sp1 binding abolished HGF-mediated SnoN induction. Furthermore, HGF selectively induced CREB phosphorylation in HKC-8 cells but not in mesangial cells and fibroblasts. In vivo, administration of HGF gene induced renal Erk-1/2 phosphorylation, CREB activation, and SnoN expression in obstructive nephropathy. Collectively, these results suggest that CREB activation, in concert with Sp1, constitutes a molecular switch that confers the cell type-specific induction of SnoN in response to HGF stimulation.
Collapse
Affiliation(s)
- Ruoyun Tan
- Department of Pathology, University of Pittsburgh, S-405 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
32
|
Isobe M, Futamatsu H, Suzuki JI. Hepatocyte growth factor: Effects on immune-mediated heart diseases. Trends Cardiovasc Med 2006; 16:188-93. [PMID: 16839861 DOI: 10.1016/j.tcm.2006.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2006] [Revised: 03/09/2006] [Accepted: 03/13/2006] [Indexed: 10/24/2022]
Abstract
There is growing evidence of the potential role of hepatocyte growth factor (HGF) in various cardiovascular diseases. In addition to the beneficial effects of HGF in myocardial infarction, heart failure, and occlusive peripheral arterial disease, administration of HGF effectively suppresses acute and chronic cardiac allograft rejection and autoimmune myocarditis. The present review summarizes recent advances in the utility of HGF for heart diseases, especially immune-mediated heart diseases. Possible mechanisms of action in the suppression of T-cell-mediated immunity are also discussed.
Collapse
Affiliation(s)
- Mitsuaki Isobe
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Bunkyoku, Tokyo, Japan.
| | | | | |
Collapse
|
33
|
Zhang A, Wang MH, Dong Z, Yang T. Prostaglandin E2 is a potent inhibitor of epithelial-to-mesenchymal transition: interaction with hepatocyte growth factor. Am J Physiol Renal Physiol 2006; 291:F1323-31. [PMID: 16868306 DOI: 10.1152/ajprenal.00480.2005] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) has emerged as a critical event in the pathogenesis of tubulointerstitial fibrosis. EMT is typically induced by transforming growth factor-beta1 (TGF-beta1) and inhibited by hepatocyte growth factor (HGF). The present study was undertaken to evaluate the potential role of cyclooxygenase (COX)-2-derived PGE2 in regulation of EMT in cultured Madin-Darby canine kidney (MDCK) cells, in the setting of HGF treatment. Exposure to 50 ng/ml HGF significantly induced COX-2 protein expression and PGE2 release, whereas other growth factors, including epidermal growth factor, the insulin-like growth factor I protein, platelet-derived growth factor-BB, and TGF-beta1, had no effects on COX-2 expression or PGE2 release. COX-2 induction by HGF was preceded by activation of ERK1/2, and an ERK1/2-specific inhibitor, U-0126 (10 microM), completely abolished HGF-induced COX-2 expression. Exposure of MDCK cells to 10 ng/ml TGF-beta1 for 72 h induced EMT as evidenced by conversion to the spindle-like morphology, loss of E-cadherin, and activation of alpha-smooth muscle actin. In contrast, treatment with 1 microM PGE2 completely blocked EMT, associated with a significant elevation of intracellular cAMP and complete blockade of TGF-beta1-induced oxidant production. cAMP-elevating agents, including 8-Br-cAMP, forskolin, and IBMX, inhibited EMT and associated oxidative stress induced by TGF-beta1, but inhibition of cAMP pathway with Rp-cAMP, the cAMP analog, and H89, the protein kinase A (PKA) inhibitor, did not block the effect of PGE2. The effect of HGF on EMT was inhibited by approximately 50% in the presence of a COX-2 inhibitor SC-58635 (10 microM). Therefore, our data suggest that PGE2 inhibits EMT via inhibition of oxidant production and COX-2-derived PGE2 partially accounts for the antifibrotic effect of HGF.
Collapse
Affiliation(s)
- Aihua Zhang
- Division of Nephrology, University of Utah and VA Medical Center, Salt Lake City, UT 84148, USA
| | | | | | | |
Collapse
|
34
|
Abstract
Hepatocyte growth factor (HGF) has emerged as a potent, endogenous antifibrotic factor that shows an impressive efficacy in ameliorating tissue fibrosis in a wide variety of animal models. Herrero-Fresneda et al. provide new evidence demonstrating that intramuscular injection of HGF gene reduces mortality, inflammation, and renal fibrosis in chronic allograft nephropathy.
Collapse
Affiliation(s)
- Y Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA.
| | | |
Collapse
|
35
|
Wada T, Azuma H, Furuichi K, Sakai N, Kitagawa K, Iwata Y, Matsushima K, Takahara S, Yokoyama H, Kaneko S. Reduction in chronic allograft nephropathy by inhibition of p38 mitogen-activated protein kinase. Am J Nephrol 2006; 26:319-25. [PMID: 16816533 DOI: 10.1159/000094365] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Accepted: 06/05/2006] [Indexed: 01/03/2023]
Abstract
BACKGROUND Chronic allograft nephropathy (CAN) is the major cause for late graft loss and is therefore a key target for therapy. METHODS The impact of p38 mitogen-activated kinase (MAPK) on CAN was investigated by administering FR167653 (32 mg/kg/day), a specific inhibitor of p38 MAPK, for 4 weeks in addition to conventional cyclosporine therapy (1.5 mg/kg/day for 5 days) in an established experimental rat transplantation model. RESULTS Transplanted rats develop glomerulosclerosis, arterial obliteration, interstitial fibrosis and tubular atrophy, all of which are characteristic of CAN, resulting in shortened survival on 32 weeks. However, the inhibition of p38 MAPK by daily subcutaneous treatment with FR167653 resulted in reduced CAN with preserved renal function and prolonged survival. The FR167653-treated rats had fewer phosphorylated p38 MAPK-positive cells in treated kidneys. Concomitantly, the expression of monocyte chemoattractant protein-1/CCL2 and transforming growth factor-beta(1) was markedly reduced. CONCLUSION These results suggest that p38 MAPK phosphorylation is involved in the pathogenesis of CAN and provide evidence that p38 MAPK is a novel, appealing therapeutic target for combating CAN.
Collapse
Affiliation(s)
- Takashi Wada
- Department of Gastroenterology and Nephrology, Graduate School of Medical Science, Kanazawa University, Takara-machi, Kanazawa, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Herrero-Fresneda I, Torras J, Franquesa M, Vidal A, Cruzado JM, Lloberas N, Fillat C, Grinyó JM. HGF gene therapy attenuates renal allograft scarring by preventing the profibrotic inflammatory-induced mechanisms. Kidney Int 2006; 70:265-74. [PMID: 16710352 DOI: 10.1038/sj.ki.5001510] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammatory processes and tissue scarring are characteristic features of chronic allograft nephropathy. Hepatocyte growth factor (HGF) has beneficial effects on renal fibrosis and it also ameliorates renal interstitial inflammation as it has been recently described. Contrarily to protein administration, intramuscular gene electrotransfer allows sustained release of HGF. So, here we hypothesized that gene therapy with human HGF would diminish the characteristic scarring of chronic allograft nephropathy either by antagonizing tissue fibrosis mechanisms or by reducing inflammation. Lewis rats transplanted with cold preserved Fischer kidneys received vehicle (NoHGF) or intramuscular plasmid DNA encoding HGF plus electroporation either before transplantation (IniHGF, early post-transplant cytoprotection of tubular cells) or 8/10 weeks after transplantation (DelHGF, delayed prevention of chronic mechanisms). Serum creatinine and proteinuria were measured every 4 weeks for 24 weeks. Grafts at 12 or 24 weeks were evaluated for glomerulosclerosis, fibrosis inflammatory cells and mediators, cell regeneration and tubulo-interstitial damage. Nontreated animals developed renal insufficiency, progressive proteinuria and fibrosis among other characteristic histological features of chronic allograft nephropathy. Treatment with human HGF, especially when delayed until the onset of fibrogenic mechanisms, reduced renal failure and mortality, diminished tubule-interstitial damage, induced cell regeneration, decreased inflammation, NF-kappaB activation, and profibrotic markers at 12 weeks and prevented late interstitial fibrosis and glomerulosclerosis. The effectiveness of HGF-gene therapy in the prevention of renal allograft scarring is related with the halt of profibrotic inflammatory-induced mechanisms.
Collapse
Affiliation(s)
- I Herrero-Fresneda
- Laboratory of Experimental Nephrology, Department of Medicine, IDIBELL-Hospital Universitari de Bellvitge, Barcelona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The dramatic improvements in short-term graft survival and acute rejection rates could only have been dreamed of 20 years ago. Late graft loss following kidney transplantation is now the critical issue of this decade. Frequently, graft loss is associated with the development of tubular atrophy and interstitial fibrosis within the kidney (i.e. chronic allograft nephropathy; CAN). Major treatment strategies in this disorder are non-specific and the focus of intervention has been on limiting injurious events. Following graft injury is a fibrogenesis phase featuring both proliferative and infiltrative responses mediated by chemokines, cytokines and growth factors. In particular, TGFbeta has been strongly implicated in the pathogenesis of chronic injury and epithelial-mesenchymal transformation (EMT) may be part of this process. The cascade of events results in matrix accumulation, due to either increased production and/or reduced degradation of matrix. Recent investigations into the pathogenesis of tissue fibrosis have suggested a number of new strategies to ameliorate matrix synthesis. While the majority of therapies have focused on TGFbeta, this may not be an ideal maneuver in transplant settings and alternative targets identified in other fibrotic diseases will be discussed. Attacking graft fibrosis should be a new focus in organ transplantation.
Collapse
Affiliation(s)
- R B Mannon
- Transplantation Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
38
|
Tsau YK, Tsai IJ, Chen YM. Transient reciprocal change of renal hepatocyte growth factor and transforming growth factor-beta1 may relate to renal hypertrophy in rats with liver injury or unilateral nephrectomy. Pediatr Res 2006; 59:494-9. [PMID: 16549518 DOI: 10.1203/01.pdr.0000203101.18174.fe] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We examined an animal model of liver injury using ligation of the common bile duct (CBD) in young rats to observe nephromegaly and to determine plasma and renal changes in hepatocyte growth factor (HGF) and transforming growth factor (TGF)-beta1. To examine the role of TGF-beta1 in the process of compensatory renal growth, renal tissue HGF, TGF-beta1, TGF-beta1 mRNA, and c-met protein were measured. Plasma HGF level decreased significantly at 1 wk, and plasma TGF-beta1 level also decreased at 1 wk and remained low at 2 wk after surgery in CBD ligation rats. Increased renal HGF/TGF-beta1 ratio was noted at 2 wk, followed by a higher kidney weight/body weight ratio and an elevated protein/DNA ratio at 3 wk after operation in CBD ligation rats. The increased renal HGF/TGF-beta1 ratio in CBD ligation rats was mainly attributed to elevated renal HGF levels. Renal HGF/TGF-beta1 ratio was also elevated at 12 h after unilateral nephrectomy. This elevated renal HGF/TGF-beta1 resulted exclusively from low renal TGF-beta1. Renal TGF-beta1 mRNA decreased significantly at 12-24 h after surgery in unilateral nephrectomized rats, whereas renal c-met receptor protein levels increased. Transient reciprocal change of HGF and TGF-beta1 manifesting as an increased renal HGF/TGF-beta1 ratio soon after uninephrectomy and later during CBD ligation suggests the probable role of TGF-beta1 in renal growth control and its possible initiating of renal hypertrophy in liver injury or unilateral nephrectomy.
Collapse
Affiliation(s)
- Yong-Kwei Tsau
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| | | | | |
Collapse
|
39
|
Barry FP, Murphy JM, English K, Mahon BP. Immunogenicity of adult mesenchymal stem cells: lessons from the fetal allograft. Stem Cells Dev 2005; 14:252-65. [PMID: 15969620 DOI: 10.1089/scd.2005.14.252] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Herein we review recent data that support host tolerance of allogeneic adult mesenchymal stem cells (MSC). Evidence is emerging that donor MSC deploy a very powerful array of mechanisms that allow escape from host allogeneic responses. These mechanisms include limited expression of alloantigen by the stem cell and cell contact-dependent and -independent mechanisms. MSC modulate host dendritic cell and T cell function, promoting induction of suppressor or regulatory T cells. These effects are complemented by the induction of divisional arrest anergy in T cells and by stem cell production of soluble immunomodulatory factors, including interleukin-10, transforming growth factor-beta, prostaglandin E2, and hepatocyte growth factor. In addition, MSC express the enzyme indoleamine 2,3-dioxygenase, which creates a tryptophan-depleted milieu that promotes immunosuppression. We propose that these observations show striking similarity to emerging data on the maternal acceptance of the fetal allograft. This comparison suggests new approaches to determine the contribution of different mechanisms to the successful use of MSC in regenerative medicine.
Collapse
Affiliation(s)
- Frank P Barry
- Regenerative Medicine Institute (REMEDI), National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | | | | | | |
Collapse
|
40
|
Ryan JM, Barry FP, Murphy JM, Mahon BP. Mesenchymal stem cells avoid allogeneic rejection. JOURNAL OF INFLAMMATION-LONDON 2005; 2:8. [PMID: 16045800 PMCID: PMC1215510 DOI: 10.1186/1476-9255-2-8] [Citation(s) in RCA: 638] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2005] [Accepted: 07/26/2005] [Indexed: 02/07/2023]
Abstract
Adult bone marrow derived mesenchymal stem cells offer the potential to open a new frontier in medicine. Regenerative medicine aims to replace effete cells in a broad range of conditions associated with damaged cartilage, bone, muscle, tendon and ligament. However the normal process of immune rejection of mismatched allogeneic tissue would appear to prevent the realisation of such ambitions. In fact mesenchymal stem cells avoid allogeneic rejection in humans and in animal models. These finding are supported by in vitro co-culture studies. Three broad mechanisms contribute to this effect. Firstly, mesenchymal stem cells are hypoimmunogenic, often lacking MHC-II and costimulatory molecule expression. Secondly, these stem cells prevent T cell responses indirectly through modulation of dendritic cells and directly by disrupting NK as well as CD8+ and CD4+ T cell function. Thirdly, mesenchymal stem cells induce a suppressive local microenvironment through the production of prostaglandins and interleukin-10 as well as by the expression of indoleamine 2,3,-dioxygenase, which depletes the local milieu of tryptophan. Comparison is made to maternal tolerance of the fetal allograft, and contrasted with the immune evasion mechanisms of tumor cells. Mesenchymal stem cells are a highly regulated self-renewing population of cells with potent mechanisms to avoid allogeneic rejection.
Collapse
Affiliation(s)
- Jennifer M Ryan
- Institute of Immunology, National University of Ireland, Maynooth, Co. Kildare Ireland
| | - Frank P Barry
- Regenerative Medicine Institute (REMEDI), National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - J Mary Murphy
- Regenerative Medicine Institute (REMEDI), National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Bernard P Mahon
- Institute of Immunology, National University of Ireland, Maynooth, Co. Kildare Ireland
| |
Collapse
|
41
|
Mizuno S, Nakamura T. Prevention of neutrophil extravasation by hepatocyte growth factor leads to attenuations of tubular apoptosis and renal dysfunction in mouse ischemic kidneys. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1895-905. [PMID: 15920173 PMCID: PMC1602426 DOI: 10.1016/s0002-9440(10)62498-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ischemia and reperfusion (I/R) injuries occur in numerous organs under pathophysiological conditions. In this process, neutrophils play important roles in eliciting parenchymal injuries. Using a murine model of renal I/R, we show that hepatocyte growth factor (HGF) is a natural ligand that inhibits endothelial injuries and neutrophil extravasation. In mice after renal I/R, plasma HGF levels increased, along with c-Met/HGF receptor phosphorylation in the vascular endothelium. However, this c-Met activation was transient, associated with a decrease in endogenous HGF level, and followed by neutrophil infiltration and renal dysfunction. Suppression of endothelial c-Met phosphorylation by anti-HGF IgG led to rapid progressions of neutrophil extravasation, tubular apoptosis, and renal dysfunction. Inversely, enhancement of the c-Met activation by exogenous HGF blocked endothelial/tubular apoptotic injuries and acute renal failure. In this process, HGF prevented endothelial nuclear factor kappaB activation and inhibited induction of an adhesion molecule (ICAM-1), resulting in attenuated vascular edema and neutrophil infiltration. Thus, we conclude that 1) the HGF/c-Met system of endothelial cells confers an initial barrier to block neutrophil infiltration, and 2) transient and insufficient HGF production allows manifestation of postischemic renal failure. Our study provides a rationale for why HGF supplementation elicits therapeutic effects in ischemic kidneys.
Collapse
Affiliation(s)
- Shinya Mizuno
- Division of Molecular Regenerative Medicine, Department of Regenerative Medicine, Course of Advanced Medicine, Osaka University Graduate School of Medicine, 2-2-B7 Yamadaoka, Suita 565-0871, Japan
| | | |
Collapse
|
42
|
Isaka Y, Yamada K, Takabatake Y, Mizui M, Miura-Tsujie M, Ichimaru N, Yazawa K, Utsugi R, Okuyama A, Hori M, Imai E, Takahara S. Electroporation-mediated HGF gene transfection protected the kidney against graft injury. Gene Ther 2005; 12:815-20. [PMID: 15772690 DOI: 10.1038/sj.gt.3302478] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The annual rate of kidney graft loss caused by chronic allograft nephropathy (CAN) has not improved over the past decade. Recent reports suggest that acute renal ischemia results in development of CAN. The goal of the present study was to assess the renoprotective potential and safety of hepatocyte growth factor (HGF) gene transfer using a porcine kidney transplant warm ischemia injury model. Following left porcine kidney removal, 10 min of warm ischemic injury was intentionally induced. Next, the HGF expression vector or vehicle was infused into the renal artery with the renal vein clamped ex vivo, and electric pulses were discharged using bathtub-type electrodes. Kidney grafts were then transplanted after removing the right kidney. Histopathological examination of vehicle-transfected kidney transplant revealed initial tubular injury followed by tubulointerstitial fibrosis. In contrast, HGF-transfected kidneys showed no initial tubular damage and no interstitial fibrosis at 6 months post-transplant. We conclude that electroporation-mediated ex vivo HGF gene transfection protects the kidney against graft injury in a porcine model.
Collapse
Affiliation(s)
- Y Isaka
- Department of Internal Medicine and Therapeutics, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Herrero-Fresneda I, Torras J, Vidal A, Lloberas N, Cruzado JM, Grinyó JM. Reduction of postischemic immune inflammatory response: an effective strategy for attenuating chronic allograft nephropathy. Transplantation 2005; 79:165-73. [PMID: 15665764 DOI: 10.1097/01.tp.0000147198.88801.57] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Ischemia added to the allogeneic background accelerates the cellular mechanisms involved in alloresponsiveness, supporting the influence of early nonspecific inflammatory injury on chronic allograft nephropathy (CAN). The authors hypothesize that reinforcing initial immunosuppressive regimens may prevent immunogenicity derived from postischemic inflammatory responses, attenuating CAN. METHODS Lewis rats engrafted with Fischer kidneys received for 15 days overimmunosuppressive doses of rapamycin, a standard cyclosporine regimen, or both, and were followed functionally for 24 weeks. Animals were grouped according to the initial immunosuppressant or cold-ischemia period. Grafts were evaluated for acute inflammatory response at 1 week and for chronic histologic damage at 24 weeks. RESULTS Rats under cyclosporine alone displayed the highest mortality, which was decreased in the long term by reducing cold ischemia or by strengthening immunosuppression. At 24 weeks, all rapamycin-treated groups displayed much less severe tubulointerstitial and vascular damage. The combination of both immunosuppressants offered better functional outcome and a global reduction in chronic histologic damage. After 1 week, ATN and profibrotic features appeared in all 5-hr ischemic animals, indicating that cyclosporine and rapamycin co-treatment did not induce further nephrotoxicity. Treatment with rapamycin, alone or combined with cyclosporine, greatly reduced the severe immune-inflammatory damage, including vessels, shown in cyclosporine-treated ischemic grafts. CONCLUSIONS Strengthening initial immunosuppression attenuates the intensity and extent of the early postischemic immune-inflammatory response as well as later function and structure of renal allografts. Severe CAN may be prevented by reducing cold ischemia or strengthening immunosuppression. Because the former approach is not always possible, reinforcement of early immunosuppression constitutes an excellent alternative.
Collapse
Affiliation(s)
- Immaculada Herrero-Fresneda
- Laboratory of Experimental Nephrology, Department of Medicine, Universitat de Barcelona, Hospital Universitari de Bellvitge, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
44
|
Yang J, Dai C, Liu Y. A novel mechanism by which hepatocyte growth factor blocks tubular epithelial to mesenchymal transition. J Am Soc Nephrol 2005; 16:68-78. [PMID: 15537870 DOI: 10.1681/asn.2003090795] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hepatocyte growth factor (HGF) is a potent antifibrotic cytokine that blocks tubular epithelial to mesenchymal transition (EMT) induced by TGF-beta1. However, the underlying mechanism remains largely unknown. This study investigated the signaling events that lead to HGF blockade of the TGF-beta1-initiated EMT. Incubation of human kidney epithelial cells HKC with HGF only marginally affected the expression of TGF-beta1 and its type I and type II receptors, suggesting that disruption of TGF-beta1 signaling likely plays a critical role in mediating HGF inhibition of TGF-beta1 action. However, HGF neither affected TGF-beta1-induced Smad-2 phosphorylation and its subsequent nuclear translocation nor influenced the expression of inhibitory Smad-6 and -7 in tubular epithelial cells. HGF specifically induced the expression of Smad transcriptional co-repressor SnoN but not Ski and TG-interacting factor at both mRNA and protein levels in HKC cells. SnoN physically interacted with activated Smad-2 by forming transcriptionally inactive complex and overrode the profibrotic action of TGF-beta1. In vivo, HGF did not affect Smad-2 activation and its nuclear accumulation in tubular epithelium, but it restored SnoN protein abundance in the fibrotic kidney in obstructive nephropathy. Hence, HGF blocks EMT by antagonizing TGF-beta1's action via upregulating Smad transcriptional co-repressor SnoN expression. These findings not only identify a novel mode of interaction between the signals activated by HGF receptor tyrosine kinase and TGF-beta receptor serine/threonine kinases but also illustrate the feasibility of confining Smad activity as an effective strategy for blocking renal fibrosis.
Collapse
Affiliation(s)
- Junwei Yang
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Pittsburgh School of Medicine, S-405 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
45
|
Yamaura K, Ito KI, Tsukioka K, Wada Y, Makiuchi A, Sakaguchi M, Akashima T, Fujimori M, Sawa Y, Morishita R, Matsumoto K, Nakamura T, Suzuki JI, Amano J, Isobe M. Suppression of Acute and Chronic Rejection by Hepatocyte Growth Factor in a Murine Model of Cardiac Transplantation. Circulation 2004; 110:1650-7. [PMID: 15364799 DOI: 10.1161/01.cir.0000143052.45956.71] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background—
Although treatment with immunosuppressive agents has contributed to overcoming acute rejection and improving the midterm survival of transplanted hearts, cardiac allograft vasculopathy (CAV) has remained the main cause of primary graft failure. Recent approaches have shown that hepatocyte growth factor (HGF) exhibits cardiotrophic functions. We therefore addressed whether HGF would regulate acute and chronic rejection in cardiac transplantation.
Methods and Results—
We used a murine heterotopic cardiac transplantation model between fully incompatible strains and administered 500 μg · kg
−1
· d
−1
HGF during the initial 14 days after transplantation. The HGF-treated allografts showed significantly prolonged survival (42.3±4.1 days,
P
<0.001) compared with the controls (11.1±0.6 days), with tolerance induction in 47.4%. Histopathologically, the number of infiltrating cells was significantly decreased and myocardial necrosis was less prominent with a reduction of apoptosis in the allografts by HGF treatment during acute rejection. In the long-term surviving allografts, HGF significantly inhibited the development of CAV and interstitial fibrosis. With respect to intragraft cytokine mRNA expression, HGF treatment reduced the early expression of interferon-γ and enhanced the expression of transforming growth factor-β1 during the acute phase and of interleukin-10 continuously through the acute phase to the chronic phase.
Conclusions—
Our findings demonstrate that HGF can prolong the survival of allografts by its cardioprotective and immunomodulative potencies. Thus, HGF administration may constitute a new therapeutic approach to preventing cardiac graft failure that has not been overcome by conventional immunosuppressive agents.
Collapse
Affiliation(s)
- Kazuhiro Yamaura
- Department of Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Liu Y. Hepatocyte growth factor in kidney fibrosis: therapeutic potential and mechanisms of action. Am J Physiol Renal Physiol 2004; 287:F7-F16. [PMID: 15180923 DOI: 10.1152/ajprenal.00451.2003] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hepatocyte growth factor (HGF) is a pleiotropic factor that plays an imperative role in tubular repair and regeneration after acute renal injury. Growing evidence indicates that HGF is also an endogenous renoprotective factor that possesses a potent antifibrotic ability. HGF prevents the initiation and progression of chronic renal fibrosis and inhibits transforming growth factor (TGF)-beta(1) expression in a wide variety of animal models. In vitro, HGF counteracts the action of TGF-beta(1) in different types of kidney cells, resulting in blockade of the myofibroblastic activation from interstitial fibroblasts and glomerular mesangial cells, as well as inhibition of the mesenchymal transition from tubular epithelial cells. Recent studies reveal that HGF antagonizes the profibrotic actions of TGF-beta(1) by intercepting Smad signal transduction through diverse mechanisms. In interstitial fibroblasts, HGF blocks activated Smad-2/3 nuclear translocation, whereas it specifically upregulates the expression of the Smad transcriptional corepressor SnoN in tubular epithelial cells. In glomerular mesangial cells, HGF stabilizes another Smad corepressor, TGIF, by preventing it from degradation. Smad corepressors bind to activated Smad-2/3 and sequester their ability to transcriptionally activate TGF-beta target genes. This article reviews recent advances in our understanding of the cellular and molecular mechanisms underlying HGF inhibition of renal fibrosis.
Collapse
Affiliation(s)
- Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
47
|
Abstract
Kidney-targeted gene therapy could be an ideal treatment for renal diseases since the therapeutic molecule is limited in the kidney and the systemic effect may be minimized. The technical development of the gene delivery to kidney and the identification of the responsive gene for a particular disease encourage the challenge to hereditary diseases. Collagen type IV reassembling was reported to be succeeded in Alport syndrome model by introduction of exogenous COL4A5 gene. Many gene therapies are evaluated in various glomerulonephritis models and unilateral ureteral obstruction (UUO) model, and favorable results are accumulated. Transplant kidney is an ideal target for gene therapy, by which ischemia reperfusion, acute rejection and chronic allograft nephropathy can be treated. The importation of the novel technology, for example hybrid stem cell-gene therapy could promote the gene therapy of renal diseases toward clinical application.
Collapse
Affiliation(s)
- Enyu Imai
- Division of Nephrology, Department of Internal Medicine, Osaka University Graduate School of Medicine, Japan.
| | | | | | | |
Collapse
|
48
|
Cruzado JM, Lloberas N, Torras J, Riera M, Fillat C, Herrero-Fresneda I, Aran JM, Alperovich G, Vidal A, Grinyó JM. Regression of advanced diabetic nephropathy by hepatocyte growth factor gene therapy in rats. Diabetes 2004; 53:1119-27. [PMID: 15047630 DOI: 10.2337/diabetes.53.4.1119] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Diabetic nephropathy is the main cause of end-stage renal disease requiring dialysis in developed countries. In this study, we demonstrated the therapeutic effect of hepatocyte growth factor (HGF) on advanced rather than early diabetic nephropathy using a rat model of streptozotocin-induced diabetes. Early diabetic nephropathy (16 weeks after induction of diabetes) was characterized by albuminuria, hyperfiltration, and glomerular hypertrophy, whereas advanced diabetic nephropathy showed prominent transforming growth factor (TGF)-beta1 upregulation, mesangial expansion, and glomerulosclerosis. An SP1017-formulated human HGF (hHGF) plasmid was administered by intramuscular injection combined with electroporation over a 30-day follow-up in rats with early and advanced diabetic nephropathy. hHGF gene therapy upregulated endogenous rat HGF in the diabetic kidney (rat HGF by RT-PCR was threefold higher than in diabetic rats without therapy). hHGF gene therapy did not improve functional or morphologic abnormalities in early diabetic nephropathy. hHGF gene therapy reduced albuminuria and induced strong regression of mesangial expansion and glomerulosclerosis in advanced diabetic nephropathy. These findings were associated with suppression of renal TGF-beta1 and mesangial connective tissue growth factor (CTGF) upregulation, inhibition of renal tissue inhibitor of metalloproteinase (TIMP)-1 expression, and reduction of renal interstitial myofibroblasts. In conclusion, our results suggest that hHGF gene therapy may be considered as an innovative therapeutic strategy to treat advanced diabetic nephropathy.
Collapse
Affiliation(s)
- Josep M Cruzado
- Nephrology Service, Medicine Department, Hospital Universitari Bellvitge, University of Barcelona, Catalonia, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Somatic cell gene therapy has made considerable progress last five years and has shown clear success in some clinical trials. In the field of nephrology, both the elucidation of pathophysiology of renal diseases and the development of gene transfer technique have become driving force for new therapy of incurable renal diseases, such as Alport syndrome and polycystic kidney disease. Gene therapy of renal cancer, although its application is limited to advanced cancer, is the front-runner of clinical application. Erythropoietin gene therapy has provided encouraging results for the treatment of anemia in uremic rats and recently progressed to the inducible one in response to hypoxia. Gene therapy for glomerulonephritis and renal fibrosis showed prominent impact on experimental models, although the safety must be confirmed for prolonged treatment. Transplant kidney is an ideal material for gene modification and induction of tolerance in the transplant kidney is an attractive challenge. Emerging techniques are becoming available such as stem cell technology and messenger RNA silencing strategies. We believe that the future of gene therapy research is exciting and promising and it holds an enormous potential for clinical application.
Collapse
Affiliation(s)
- Enyu Imai
- Division of Nephrology, Department of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871
| | | |
Collapse
|
50
|
Mizuno S, Nakamura T. Suppressions of chronic glomerular injuries and TGF-β1production by HGF in attenuation of murine diabetic nephropathy. Am J Physiol Renal Physiol 2004; 286:F134-43. [PMID: 14519594 DOI: 10.1152/ajprenal.00199.2003] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diabetic nephropathy is now the leading cause of end-stage renal diseases, and glomerular sclerotic injury is an initial event that provokes renal dysfunction during processes of diabetes-linked kidney disease. Growing evidence shows that transforming growth factor-β1(TGF-β1) plays a key role in this process, especially in eliciting hypertrophy and matrix overaccumulation. Thus it is important to find a ligand system to antagonize the TGF-β1-mediated pathogenesis under high-glucose conditions. Herein, we provide evidence that hepatocyte growth factor (HGF) targets mesangial cells, suppresses TGF-β1production, and minimizes glomerular sclerotic changes, using streptozotocin-induced diabetic mice. In our murine model, glomerular sclerogenesis (such as tuft area expansion and collagen deposition) progressed between 6 and 10 wk after the induction of hyperglycemia, during a natural course of diabetic disease. Glomerular HGF expression levels in the diabetic kidney transiently increased but then declined below a basal level, with manifestation of glomerular sclerogenesis. When anti-HGF IgG was injected into mice for 2 wk (i.e., from weeks 4 to 6 after onset of hyperglycemia), these glomerular changes were significantly aggravated. When recombinant HGF was injected into the mice for 4 wk (i.e., between 6 and 10 wk following streptozotocin treatment), the progression of glomerular hypertrophy and sclerosis was almost completely inhibited, even though glucose levels remained unchanged (>500 mg/dl). Even more important, HGF repressed TGF-β1production in glomerular mesangial cells even under hyperglycemic conditions both in vitro and in vivo. Consequently, not only albuminuria but also tubulointerstitial fibrogenesis were attenuated by HGF. Overall, HGF therapy inhibited the onset of renal dysfunction in the diabetic mice. On the basis of these findings, we wish to emphasize that HGF plays physiological and therapeutic roles in blocking renal fibrogenesis during a course of diabetic nephropathy.
Collapse
Affiliation(s)
- Shinya Mizuno
- Division of Molecular Regenerative Medicine, Department of Molecular Regenerative Medicine, Osaka University Graduate School of Medicine, Yamadaoka 2-2-B7, Suita 565-0871, Japan
| | | |
Collapse
|