1
|
Shakour N, Karami S, Iranshahi M, Butler AE, Sahebkar A. Antifibrotic effects of sodium-glucose cotransporter-2 inhibitors: A comprehensive review. Diabetes Metab Syndr 2024; 18:102934. [PMID: 38154403 DOI: 10.1016/j.dsx.2023.102934] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/25/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND AND AIMS Scar tissue accumulation in organs is the underlying cause of many fibrotic diseases. Due to the extensive array of organs affected, the long-term nature of fibrotic processes and the large number of people who suffer from the negative impact of these diseases, they constitute a serious health problem for modern medicine and a huge economic burden on society. Sodium-glucose cotransporter-2 inhibitors (SGLT2is) are a relatively new class of anti-diabetic pharmaceuticals that offer additional benefits over and above their glucose-lowering properties; these medications modulate a variety of diseases, including fibrosis. Herein, we have collated and analyzed all available research on SGLT2is and their effects on organ fibrosis, together with providing a proposed explanation as to the underlying mechanisms. METHODS PubMed, ScienceDirect, Google Scholar and Scopus were searched spanning the period from 2012 until April 2023 to find relevant articles describing the antifibrotic effects of SGLT2is. RESULTS The majority of reports have shown that SGLT2is are protective against lung, liver, heart and kidney fibrosis as well as arterial stiffness. According to the results of clinical trials and animal studies, many SGLT2 inhibitors are promising candidates for the treatment of fibrosis. Recent studies have demonstrated that SGLT2is affect an array of cellular processes, including hypoxia, inflammation, oxidative stress, the renin-angiotensin system and metabolic activities, all of which have been linked to fibrosis. CONCLUSION Extensive evidence indicates that SGLT2is are promising treatments for fibrosis, demonstrating protective effects in various organs and influencing key cellular processes linked to fibrosis.
Collapse
Affiliation(s)
- Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Karami
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Glucosidase inhibitor, Nimbidiol ameliorates renal fibrosis and dysfunction in type-1 diabetes. Sci Rep 2022; 12:21707. [PMID: 36522378 PMCID: PMC9755213 DOI: 10.1038/s41598-022-25848-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Diabetic nephropathy is characterized by excessive accumulation of extracellular matrix (ECM) leading to renal fibrosis, progressive deterioration of renal function, and eventually to end stage renal disease. Matrix metalloproteinases (MMPs) are known to regulate synthesis and degradation of the ECM. Earlier, we demonstrated that imbalanced MMPs promote adverse ECM remodeling leading to renal fibrosis in type-1 diabetes. Moreover, elevated macrophage infiltration, pro-inflammatory cytokines and epithelial‒mesenchymal transition (EMT) are known to contribute to the renal fibrosis. Various bioactive compounds derived from the medicinal plant, Azadirachta indica (neem) are shown to regulate inflammation and ECM proteins in different diseases. Nimbidiol is a neem-derived diterpenoid that is considered as a potential anti-diabetic compound due to its glucosidase inhibitory properties. We investigated whether Nimbidiol mitigates adverse ECM accumulation and renal fibrosis to improve kidney function in type-1 diabetes and the underlying mechanism. Wild-type (C57BL/6J) and type-1 diabetic (C57BL/6-Ins2Akita/J) mice were treated either with saline or with Nimbidiol (0.40 mg kg-1 d-1) for eight weeks. Diabetic kidney showed increased accumulation of M1 macrophages, elevated pro-inflammatory cytokines and EMT. In addition, upregulated MMP-9 and MMP-13, excessive collagen deposition in the glomerular and tubulointerstitial regions, and degradation of vascular elastin resulted to renal fibrosis in the Akita mice. These pathological changes in the diabetic mice were associated with functional impairments that include elevated resistive index and reduced blood flow in the renal cortex, and decreased glomerular filtration rate. Furthermore, TGF-β1, p-Smad2/3, p-P38, p-ERK1/2 and p-JNK were upregulated in diabetic kidney compared to WT mice. Treatment with Nimbidiol reversed the changes to alleviate inflammation, ECM accumulation and fibrosis and thus, improved renal function in Akita mice. Together, our results suggest that Nimbidiol attenuates inflammation and ECM accumulation and thereby, protects kidney from fibrosis and dysfunction possibly by inhibiting TGF-β/Smad and MAPK signaling pathways in type-1 diabetes.
Collapse
|
3
|
Jin B, Zhu J, Zhou Y, Liang L, Yang Y, Xu L, Zhang T, Li P, Pan T, Guo B, Chen T, Li H. Loss of MEN1 leads to renal fibrosis and decreases HGF-Adamts5 pathway activity via an epigenetic mechanism. Clin Transl Med 2022; 12:e982. [PMID: 35968938 PMCID: PMC9377152 DOI: 10.1002/ctm2.982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/28/2022] [Accepted: 07/03/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Renal fibrosis is a serious condition that results in the development of chronic kidney diseases. The MEN1 gene is an epigenetic regulator that encodes the menin protein and its role in kidney tissue remains unclear. METHODS Kidney histology was examined on paraffin sections stained with hematoxylin-eosin staining. Masson's trichrome staining and Sirius red staining were used to analyze renal fibrosis. Gene and protein expression were determined by quantitative real-time PCR (qPCR) and Western blot, respectively. Immunohistochemistry staining in the kidney tissues from mice or patients was used to evaluate protein levels. Flow cytometry was used to analyze the cell cycle distributions and apoptosis. RNA-sequencing was performed for differential expression genes in the kidney tissues of the Men1f/f and Men1∆/∆ mice. Chromatin immunoprecipitation sequencing (ChIP-seq) was carried out for identification of menin- and H3K4me3-enriched regions within the whole genome in the mouse kidney tissue. ChIP-qPCR assays were performed for occupancy of menin and H3K4me3 at the gene promoter regions. Luciferase reporter assay was used to detect the promoter activity. The exacerbated unilateral ureteral obstruction (UUO) models in the Men1f/f and Men1∆/∆ mice were used to assess the pharmacological effects of rh-HGF on renal fibrosis. RESULTS The expression of MEN1 is reduce in kidney tissues of fibrotic mouse and human diabetic patients and treatment with fibrotic factor results in the downregulation of MEN1 expression in renal tubular epithelial cells (RTECs). Disruption of MEN1 in RTECs leads to high expression of α-SMA and Collagen 1, whereas MEN1 overexpression restrains epithelial-to-mesenchymal transition (EMT) induced by TGF-β treatment. Conditional knockout of MEN1 resulted in chronic renal fibrosis and UUO-induced tubulointerstitial fibrosis (TIF), which is associated with an increased induction of EMT, G2/M arrest and JNK signaling. Mechanistically, menin recruits and increases H3K4me3 at the promoter regions of hepatocyte growth factor (HGF) and a disintegrin and metalloproteinase with thrombospondin motifs 5 (Adamts5) genes and enhances their transcriptional activation. In the UUO mice model, exogenous HGF restored the expression of Adamts5 and ameliorated renal fibrosis induced by Men1 deficiency. CONCLUSIONS These findings demonstrate that MEN1 is an essential antifibrotic factor in renal fibrogenesis and could be a potential target for antifibrotic therapy.
Collapse
Affiliation(s)
- Bangming Jin
- Department of SurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Transformation Engineering Research Center of Chronic Disease Diagnosis and TreatmentGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Jiamei Zhu
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Transformation Engineering Research Center of Chronic Disease Diagnosis and TreatmentGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
| | - Yuxia Zhou
- Department of SurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Transformation Engineering Research Center of Chronic Disease Diagnosis and TreatmentGuizhou Medical UniversityGuiyangChina
| | - Li Liang
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Transformation Engineering Research Center of Chronic Disease Diagnosis and TreatmentGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
| | - Yunqiao Yang
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Transformation Engineering Research Center of Chronic Disease Diagnosis and TreatmentGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
| | - Lifen Xu
- Department of SurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
| | - Tuo Zhang
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Transformation Engineering Research Center of Chronic Disease Diagnosis and TreatmentGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
| | - Po Li
- Department of SurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Ting Pan
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Transformation Engineering Research Center of Chronic Disease Diagnosis and TreatmentGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
| | - Bing Guo
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
| | - Tengxiang Chen
- Department of SurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
- School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Transformation Engineering Research Center of Chronic Disease Diagnosis and TreatmentGuizhou Medical UniversityGuiyangChina
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Haiyang Li
- Department of SurgeryAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
4
|
Grimes JA, Lourenço BN, Coleman AE, Rissi DR, Schmiedt CW. MicroRNAs are differentially expressed in the serum and renal tissues of cats with experimentally induced chronic kidney disease: a preliminary study. Am J Vet Res 2022; 83:426-433. [PMID: 35239506 DOI: 10.2460/ajvr.21.08.0136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To identify differentially expressed microRNA in the serum and renal tissues of cats with experimentally induced chronic kidney disease (CKD). SAMPLE Banked renal tissues and serum from 4 cats. PROCEDURES Cats previously underwent 90-minute unilateral ischemia with delayed contralateral nephrectomy 3 months after ischemia. Tissues were collected from the contralateral kidney at the time of nephrectomy and from the ischemic kidney 6 months after nephrectomy (study end). Serum was collected prior to ischemia (baseline serum) and at study end (end point serum). Total RNA was isolated from tissues and serum, and microRNA sequencing was performed with differential expression analysis between the contralateral and ischemic kidney and baseline and end point serum. RESULTS 20 microRNAs were differentially expressed between ischemic and contralateral kidneys, and 52 microRNAs were differentially expressed between end point and baseline serum. Five microRNAs were mutually differentially expressed between ischemic and contralateral kidneys and baseline and end point serum, with 4 (mir-21, mir-146, mir-199, and mir-235) having increased expression in both the ischemic kidney and end point serum and 1 (mir-382) having increased expression in the ischemic kidney and decreased expression in end point serum. Predicted target search for these microRNA revealed multiple genes previously shown to be involved in the pathogenesis of feline CKD, including hypoxia-inducible factor-1α, transforming growth factor-β, hepatocyte growth factor, fibronectin, and vascular endothelial growth factor A. CLINICAL RELEVANCE MicroRNAs were differentially expressed after CKD induction in this preliminary study. Regulation of renal fibrosis in feline CKD may occur through microRNA regulation of mRNAs of pro- and anti-fibrotic genes.
Collapse
Affiliation(s)
- Janet A Grimes
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Bianca N Lourenço
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Amanda E Coleman
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Daniel R Rissi
- Athens Veterinary Diagnostic Laboratory, Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Chad W Schmiedt
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA
| |
Collapse
|
5
|
Kim JH, Yang HJ, Choi SS, Kim SU, Lee HJ, Song YS. Improved bladder contractility after transplantation of human mesenchymal stem cells overexpressing hepatocyte growth factor into underactive bladder from bladder outlet obstruction models of rats. PLoS One 2021; 16:e0261402. [PMID: 34936660 PMCID: PMC8694482 DOI: 10.1371/journal.pone.0261402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/01/2021] [Indexed: 12/05/2022] Open
Abstract
Introduction An underactive bladder can lead to difficulty in voiding that causes incomplete emptying of the bladder, suggesting the need for a new strategy to increase bladder contractility in such patients. This study was performed to investigate whether human mesenchymal stem cells (hMSCs) were capable of restoring bladder contractility in rats with underactive bladder due to bladder outlet obstruction (BOO) and enhancing their effects by overexpressing hepatocyte growth factor (HGF) in hMSCs. Materials and methods The hMSCs were transplanted into the bladder wall of rats. Fifty female Sprague-Dawley rats at six weeks of age were divided into five groups: group 1: control; group 2: sham intervention; group 3: eight-week BOO; group 4: BOO rats transplanted with hMSCs; and group 5: BOO rats transplanted with hMSCs overexpressing HGF. Two weeks after the onset of BOO in groups 4 and 5, hMSCs were injected into the bladder wall. Cystometry evaluation was followed by Masson’s trichrome staining of bladder tissues. Realtime PCR and immunohistochemical staining were performed to determine for hypoxia, apoptosis, and angiogenesis. Results Collagen deposition of bladder increased in BOO but decreased after transplantation of hMSCs. The increased inter-contraction interval and residual urine volume after BOO was reversed after hMSCs transplantation. The decreased maximal voiding pressure after BOO was restored by hMSCs treatment. The mRNA expression of bladder collagen1 and TGF-β1 increased in BOO but decreased after hMSCs transplantation. The decrease in vWF-positive cells in the bladder following BOO was increased after hMSCs transplantation. Caspase 3 and TUNEL-positive apoptosis of bladder cells increased in BOO but decreased after transplantation of hMSCs. These effects were enhanced by overexpressing HGF in hMSCs. Conclusion Transplantation of hMSCs into bladder wall increased the number of micro-vessels, decreased collagen deposition and apoptosis of detrusor muscle, and improved bladder underactivity. The effects were enhanced by overexpressing HGF in hMSCs. Our findings suggest that the restoration of underactive bladder using hMSCs may be used to rectify micturition disorders in patients following resolution of BOO. Further studies are needed before hMSCs can be used in clinical applications.
Collapse
Affiliation(s)
- Jae Heon Kim
- Department of Urology, Soonchunhyang University School of Medicine, Seoul, Republic of Korea
| | - Hee Jo Yang
- Department of Urology, Soonchunhyang University School of Medicine, Cheonan, Republic of Korea
| | - Sung Sik Choi
- Medical Science Research Institute, Chungbuk National University, Cheong Ju, Republic of Korea
| | - Seung U. Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
| | - Hong J. Lee
- Medical Science Research Institute, Chungbuk National University, Cheong Ju, Republic of Korea
- * E-mail: (HJL); (YSS)
| | - Yun Seob Song
- Department of Urology, Soonchunhyang University School of Medicine, Seoul, Republic of Korea
- * E-mail: (HJL); (YSS)
| |
Collapse
|
6
|
Prado LG, Barbosa AS. Understanding the Renal Fibrotic Process in Leptospirosis. Int J Mol Sci 2021; 22:ijms221910779. [PMID: 34639117 PMCID: PMC8509513 DOI: 10.3390/ijms221910779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Leptospirosis is a neglected infectious disease caused by pathogenic species of the genus Leptospira. The acute disease is well-described, and, although it resembles other tropical diseases, it can be diagnosed through the use of serological and molecular methods. While the chronic renal disease, carrier state, and kidney fibrosis due to Leptospira infection in humans have been the subject of discussion by researchers, the mechanisms involved in these processes are still overlooked, and relatively little is known about the establishment and maintenance of the chronic status underlying this infectious disease. In this review, we highlight recent findings regarding the cellular communication pathways involved in the renal fibrotic process, as well as the relationship between renal fibrosis due to leptospirosis and CKD/CKDu.
Collapse
Affiliation(s)
- Luan Gavião Prado
- Laboratório de Bacteriologia, Instituto Butantan, Avenida Vital Brasil, 1500, São Paulo 05503-900, Brazil;
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Avenida Lineu Prestes 1374, São Paulo 05508-000, Brazil
| | - Angela Silva Barbosa
- Laboratório de Bacteriologia, Instituto Butantan, Avenida Vital Brasil, 1500, São Paulo 05503-900, Brazil;
- Correspondence:
| |
Collapse
|
7
|
Wang Q, Tao Y, Xie H, Liu C, Liu P. MicroRNA‑101 inhibits renal tubular epithelial‑to‑mesenchymal transition by targeting TGF‑β1 type I receptor. Int J Mol Med 2021; 47:119. [PMID: 33955520 PMCID: PMC8099196 DOI: 10.3892/ijmm.2021.4952] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 03/30/2021] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are key regulators of renal interstitial fibrosis (RIF). The present study was designed to identify miRNAs associated with the development of RIF, and to explore the ability of these identified miRNAs to modulate the renal tubular epithelial‑to‑mesenchymal transition (EMT) process. To this end, miRNAs that were differentially expressed between normal and fibrotic kidneys in a rat model of mercury chloride (HgCl2)‑induced RIF were detected via an array‑based approach. Bioinformatics analyses revealed that miR‑101 was the miRNA that was most significantly downregulated in the fibrotic renal tissue samples, and this was confirmed by RT‑qPCR, which also demonstrated that this miRNA was downregulated in transforming growth factor (TGF)‑β1‑treated human proximal tubular epithelial (HK‑2) cells. When miR‑101 was overexpressed, this was sufficient to reverse TGF‑β1‑induced EMT in HK‑2 cells, leading to the upregulation of the epithelial marker, E‑cadherin, and the downregulation of the mesenchymal marker, α‑smooth muscle actin. By contrast, the downregulation of miR‑101 using an inhibitor exerted the opposite effect. The overexpression of miR‑101 also suppressed the expression of the miR‑101 target gene, TGF‑β1 type I receptor (TβR‑I), and thereby impaired TGF‑β1/Smad3 signaling, while the opposite was observed upon miR‑101 inhibition. To further confirm the ability of miR‑101 to modulate EMT, the HK‑2 cells were treated with the TβR‑I inhibitor, SB‑431542, which significantly suppressed TGF‑β1‑induced EMT in these cells. Notably, miR‑101 inhibition exerted a less pronounced effect upon EMT‑related phenotypes in these TβR‑I inhibitor‑treated HK‑2 cells, supporting a model wherein miR‑101 inhibits TGF‑β1‑induced EMT by suppressing TβR‑I expression. On the whole, the present study demonstrates that miR‑101 is capable of inhibiting TGF‑β1‑induced tubular EMT by targeting TβR‑I, suggesting that it may be an important regulator of RIF.
Collapse
Affiliation(s)
- Qinglan Wang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yanyan Tao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Hongdong Xie
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Chenghai Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Ping Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
8
|
Lee M, Kim SH, Jhee JH, Kim TY, Choi HY, Kim HJ, Park HC. Microparticles derived from human erythropoietin mRNA-transfected mesenchymal stem cells inhibit epithelial-to-mesenchymal transition and ameliorate renal interstitial fibrosis. Stem Cell Res Ther 2020; 11:422. [PMID: 32993806 PMCID: PMC7523343 DOI: 10.1186/s13287-020-01932-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/15/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
Background Renal tubulointerstitial fibrosis (TIF) plays an important role in the progression of chronic kidney disease (CKD) and its pathogenesis involves epithelial-to-mesenchymal transition (EMT) upon renal injury. Recombinant human erythropoietin (rhEPO) has been shown to display novel cytoprotective effects, in part by inhibiting transforming growth factor (TGF)-β1-induced EMT. Here, we evaluated the inhibitory effects of microparticles (MPs) derived from human EPO gene-transfected kidney mesenchymal stem cells (hEPO-KMSCs) against TGF-β1-induced EMT in Madin-Darby canine kidney (MDCK) cells and against TIF in mouse kidneys with unilateral ureteral obstruction (UUO). Methods EMT was induced in MDCK cells by treatment with TGF-β1 (5 ng/mL) for 48 h and then inhibited by co-treatment with rhEPO (100 IU/mL), mock gene-transfected KMSC-derived MPs (MOCK-MPs), or hEPO-KMSC-derived MPs (hEPO-MPs) for a further 48 h. UUO was induced in FVB/N mice, which were then treated with rhEPO (1000 IU/kg, intraperitoneally, every other day for 1 week), MOCK-MPs, or hEPO-MPs (80 μg, intravenously). Alpha-smooth muscle actin (α-SMA), fibronectin, and E-cadherin expression were evaluated in MDCK cells and kidney tissues, and the extent of TIF in UUO kidneys was assessed by immunohistochemical staining. Results TGF-β1 treatment significantly increased α-SMA and fibronectin expression in MDCK cells and decreased that of E-cadherin, while co-treatment with rhEPO, MOCK-MPs, or hEPO-MPs markedly attenuated these changes. In addition, rhEPO and hEPO-MP treatment effectively decreased phosphorylated Smad2 and Smad3, as well as phosphorylated p38 mitogen-activated protein kinase (MAPK) expression, suggesting that rhEPO and rhEPO-MPs can inhibit TGF-β1-induced EMT via both Smad and non-Smad pathways. rhEPO and hEPO-MP treatment also significantly attenuated the extent of renal TIF after 1 week of UUO compared to MOCK-MPs, with hEPO-MPs significantly reducing myofibroblast and F4/80+ macrophage infiltration as well as EMT marker expression in UUO renal tissues in a similar manner to rhEPO. Conclusions Our results demonstrate that hEPO-MPs modulate TGF-β1-induced EMT in MDCK cells via the Smad2, Smad3, and p38 MAPK pathways and significantly attenuated renal TIF in UUO kidneys.
Collapse
Affiliation(s)
- Mirae Lee
- Graduate Program of Nano Science and Technology, Yonsei University, Seoul, Korea.,Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seok-Hyung Kim
- Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Division of Nephrology, Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Gangwon-do, Korea
| | - Jong Hyun Jhee
- Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Tae Yeon Kim
- Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hoon Young Choi
- Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
| | - Hyung Jong Kim
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, Korea
| | - Hyeong Cheon Park
- Graduate Program of Nano Science and Technology, Yonsei University, Seoul, Korea. .,Division of Nephrology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea. .,Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Hsieh YH, Syu RJ, Lee CC, Lin SH, Lee CH, Cheng CW, Tsai JP. Arecoline induces epithelial mesenchymal transition in HK2 cells by upregulating the ERK-mediated signaling pathway. ENVIRONMENTAL TOXICOLOGY 2020; 35:1007-1014. [PMID: 32441858 DOI: 10.1002/tox.22937] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/16/2020] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Arecoline, a component of betel nuts, is a known carcinogen that causes oral cancers among those who chew betel nuts. Betel nut chewing is also associated with an increased risk of chronic kidney disease (CKD), but the role of arecoline in this association is unclear. This in vitro study investigates the effects of arecoline on cultured human kidney (HK2) cells. We observed that arecoline had no effect on cell viability but increased cell migration in a dose-dependent manner. Western blot analysis showed that arecoline treatment caused a dose-dependent decrease in E-cadherin expression and dose-dependent increases in N-cadherin, vimentin, α-SMA, and collagen expression; reverse transcriptase-polymerase chain reaction analysis revealed dose-dependent increases in α-SMA and collagen mRNA. Arecoline treatment upregulated the expression of phosphorylated extracellular signal-regulated kinase through epithelial mesenchymal transition and renal fibrosis in HK2 cells. These findings demonstrate that arecoline plays a role in inducing the epithelial mesenchymal transition and fibrogenesis in renal tubule cells and suggest that arecoline promotes the progression of CKD.
Collapse
Affiliation(s)
- Yi-Hsien Hsieh
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Clinical laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ru-Jiang Syu
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Chu-Che Lee
- Department of Medicine Research, Buddhist Dalin Tzu Chi Hospital, Chiayi, Taiwan
| | - Shin-Huey Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Hsing Lee
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of China Medical University, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Wen Cheng
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Jen-Pi Tsai
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
10
|
Yang Y, Wang Y, He Z, Liu Y, Chen C, Wang Y, Wang DW, Wang H. Trimetazidine Inhibits Renal Tubular Epithelial Cells to Mesenchymal Transition in Diabetic Rats via Upregulation of Sirt1. Front Pharmacol 2020; 11:1136. [PMID: 32848753 PMCID: PMC7403491 DOI: 10.3389/fphar.2020.01136] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Trimetazidine (TMZ), as a metabolic regulator, is effective in treatment of coronary atherosclerotic heart disease with rare side effects in the clinic for long years. Interestingly, studies have shown that TMZ protects against several acute kidney injuries (AKI). However, the effect of TMZ on chronic kidney diseases (CKD) remains unknown. This study aimed to investigate the role of TMZ in diabetic nephropathy (DN) and its potential mechanisms. A rat model of DN was established in male Sprague-Dawley rats by streptozotocin (STZ) intraperitoneal injection. Experimental rats were separated into three groups: control, DN and DN + TMZ treatment. Metabolic parameters, pathological features and renal function markers were evaluated after 20 weeks of diabetes induction. In vitro experiments, the effect of TMZ on high fat and high glucose (HFG) induced or TGFβ1-induced epithelial-to-mesenchymal transition (EMT) was examined in HK-2 cells. Our results showed that TMZ could maintain renal function without affecting hemodynamic and plasma metabolic levels in diabetic rats. The effect was associated with a reversion of pathological progression of DN, especially for tubulointerstitial fibrosis. EMT is an important contributor to renal fibrosis. In this study, we investigated the role of TMZ in the process of EMT in DN. Mechanistically; TMZ attenuated HFG-induced EMT by relieving oxidative stress via deacetylation forkhead box O1 (FoxO1) in a Sirt1-dependent pathway. And it suppressed TGFβ1-induced EMT by deacetylating Smd4 in a Sirt1-dependent manner. Moreover, our study found that TMZ upregulated Sirt1 expression by increasing the expression of nicotinamide phosphoribosyl transferase (Nampt), which is a rate limiting enzyme for nicotinamide adenine dinucleotide (NAD+) generation by salvage pathway. And the increased NAD+ promoted Sirt1 expression. In conclusion, TMZ can prevent renal dysfunction and pathogenesis of tubulointerstitial fibrosis in DN, partly by inhibition of EMT via FoxO1/ROS pathway and TGFβ/Smad pathway in a Nampt/NAD+/Sirt1 dependent manner.
Collapse
Affiliation(s)
- Yong Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Zuowen He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yunchang Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Nie L, Liu Y, Zhang B, Zhao J. Application of Histone Deacetylase Inhibitors in Renal Interstitial Fibrosis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2020; 6:226-235. [PMID: 32903948 DOI: 10.1159/000505295] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Renal interstitial fibrosis is characterized by the accumulation of extracellular matrix proteins, which is a common feature of chronic kidney diseases. SUMMARY Increasing evidence has shown the aberrant expression of histone deacetylases (HDACs) in the development and progression of renal fibrosis, suggesting the possibility of utilizing HDAC inhibitor (HDACi) as therapeutics for renal fibrosis. Recent studies have successfully demonstrated the antifibrotic effects of HDACis in various animal models, which are associated with multiple signaling pathways including TGF-β signaling, EGRF signaling, signal transducer and activator of transcription 3 pathway, and JNK/Notch2 signaling. This review will focus on the utilization of HDACi as antifibrotic agents and its relative molecular mechanisms. KEY MESSAGES HDACis have shown promising results in antifibrotic therapy, and it is rational to anticipate that HDACis will improve clinical outcomes of renal fibrosis in the future.
Collapse
Affiliation(s)
- Ling Nie
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing, China
| | - Yong Liu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing, China
| | - Bo Zhang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Army Medical University (The Third Military Medical University), Chongqing, China
| |
Collapse
|
12
|
Fung WWS, Poon PYK, Ng JKC, Kwong VWK, Pang WF, Kwan BCH, Cheng PMS, Li PKT, Szeto CC. Longitudinal Changes of NF-κB Downstream Mediators and Peritoneal Transport Characteristics in Incident Peritoneal Dialysis Patients. Sci Rep 2020; 10:6440. [PMID: 32296091 PMCID: PMC7160129 DOI: 10.1038/s41598-020-63258-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/26/2020] [Indexed: 11/09/2022] Open
Abstract
The role of intra-peritoneal mediators in the regulation peritoneal transport is not completely understood. We investigate the relation between longitudinal changes in dialysis effluent level of nuclear factor kappa-B (NF-κB) downstream mediators and the change in peritoneal transport over 1 year. We studied 46 incident PD patients. Their peritoneal transport characteristics were determined after starting PD and then one year later. Concomitant dialysis effluent levels of interleukin-6 (IL-6), cyclo-oxygenase-2 (COX-2) and hepatocyte growth factor (HGF) are determined. There were significant correlations between baseline and one-year dialysis effluent IL-6 and COX-2 levels with the corresponding dialysate-to-plasma creatinine level at 4 hours (D/P4) and mass transfer area coefficient of creatinine (MTAC). After one year, patients who had peritonitis had higher dialysis effluent IL-6 (26.6 ± 17.4 vs 15.1 ± 12.3 pg/ml, p = 0.037) and COX-2 levels (4.97 ± 6.25 vs 1.60 ± 1.53 ng/ml, p = 0.007) than those without peritonitis, and the number of peritonitis episode significantly correlated with the IL-6 and COX-2 levels after one year. In contrast, dialysis effluent HGF level did not correlate with peritoneal transport. There was no difference in any mediator level between patients receiving conventional and low glucose degradation product solutions. Dialysis effluent IL-6 and COX-2 levels correlate with the concomitant D/P4 and MTAC of creatinine. IL-6 and COX-2 may contribute to the short-term regulation of peritoneal transport.
Collapse
Affiliation(s)
- Winston Wing-Shing Fung
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Peter Yam-Kau Poon
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jack Kit-Chung Ng
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Vickie Wai-Ki Kwong
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wing-Fai Pang
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Bonnie Ching-Ha Kwan
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Phyllis Mei-Shan Cheng
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Philip Kam-Tao Li
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Cheuk-Chun Szeto
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China. .,Li Ka Shing Institute of Health Sciences (LiHS), Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
13
|
Margetts PJ, Bonniaud P. Basic Mechanisms and Clinical Implications of Peritoneal Fibrosis. Perit Dial Int 2020. [DOI: 10.1177/089686080302300604] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Peter J. Margetts
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Philippe Bonniaud
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
14
|
Xu Y, Ouyang C, Lyu D, Lin Z, Zheng W, Xiao F, Xu Z, Ding L. Diabetic nephropathy execrates epithelial-to-mesenchymal transition (EMT) via miR-2467-3p/Twist1 pathway. Biomed Pharmacother 2020; 125:109920. [PMID: 32050151 DOI: 10.1016/j.biopha.2020.109920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/12/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Although diabetic nephropathy (DN) is induced by a complicate interplay of multiple factors, the underlying mechanisms remain poorly characterized, even the treatment. Herein, we show that both of DN patients and STZ-induced type 1 diabetic rat exhibit the reduction both of urinary and circulating miR-2467-3p. We identify a negative correlation between miR-2467-3p levels and renal dysfunction. Administration of miR-2467-3p prevents diabetes-induced renal dysfunction and represses renal fibrosis in STZ-induced type 1 diabetic rats. Conversely, anti-miR-2467 overexpression exacerbates renal dysfunction and fibrosis in STZ-induced rats. In diabetic condition, the reduction of miR-2467-3p promotes expression of Twist1, inducing epithelial-to-mesenchymal transition (EMT), resulting in renal fibrosis and kidney dysfunction. Together, our study presents miR-2467/Twist1/EMT as a regulatory axis of renal dysfunction in DN.
Collapse
Affiliation(s)
- Yan Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350004, China
| | - Changhan Ouyang
- Hubei Key Laboratory of Cardiovascular, Cerebrovascular and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, China
| | - Dayin Lyu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Zhangmei Lin
- School of Clinical Medicine, Fujian Medical University, Fuzhou, 350004, China
| | - Wencai Zheng
- School of Clinical Medicine, Fujian Medical University, Fuzhou, 350004, China
| | - Fan Xiao
- School of Clinical Medicine, Fujian Medical University, Fuzhou, 350004, China
| | - Zhimin Xu
- School of Clinical Medicine, Fujian Medical University, Fuzhou, 350004, China
| | - Lexi Ding
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
15
|
Xie M, Wan J, Zhang F, Zhang R, Zhou Z, You D. Influence of hepatocyte growth factor-transfected bone marrow-derived mesenchymal stem cells towards renal fibrosis in rats. Indian J Med Res 2020; 149:508-516. [PMID: 31411175 PMCID: PMC6676852 DOI: 10.4103/ijmr.ijmr_1527_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background & objectives: Hepatocyte growth factor (HGF) produced by endothelial cells, fibroblasts, fat cells and other interstitial cells, can promote angiogenesis, repair damaged tissues and resist fibrosis. Mesenchymal stem cells (MSCs) are located in bone marrow and secrete a variety of cytokines and are often used in the repair and regeneration of damaged tissues. This study was aimed to investigate the influence of HGF-transfected bone marrow-derived MSCs towards renal fibrosis in rats. Methods: The HGF gene-carrying adenoviral vector (Ad-HGF) was transfected into MSCs, and the Ad-HGF-modified MSCs were transplanted into rats with unilateral ureteral obstruction (UUO). The localization of renal transplanted cells in the frozen section was observed with fluorescence microscope. The Masson's trichrome staining was performed to observe the renal collagen deposition, and the immunohistochemistry was performed to detect the expressions of α-smooth muscle actin (α-SMA) and HGF in renal tissues. Reverse transcription (RT)-PCR was used to detect the mRNA expressions of α-SMA, HGF and fibronectin (FN). Results: Ad-HGF-modified MSCs could highly express HGF in vitro. On the post-transplantation 3rd, 7th and 14th day, the 4',6-diamidino-2-phenylindole (DAP)-labelled transplanted cells were seen inside renal tissues. Compared with UUO group, the renal collagen deposition in transplantation group was significantly reduced, and the expressions of α-SMA mRNA and protein were significantly decreased, while the expressions of HGF mRNA and protein were significantly increased, and the expression of FN mRNA was significantly decreased (P<0.001). Interpretation & conclusions: Trans-renal artery injection of HGF-modified MSCs can effectively reduce the renal interstitial fibrosis in UUO rat model.
Collapse
Affiliation(s)
- Mingbu Xie
- Department of Nephrology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Jianxin Wan
- Department of Nephrology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Fengxia Zhang
- Department of Nephrology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Ruifang Zhang
- Department of Nephrology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Zhenhuan Zhou
- Department of Nephrology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Danyou You
- Department of Nephrology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| |
Collapse
|
16
|
Diao W, Chen W, Cao W, Yuan H, Ji H, Wang T, Chen W, Zhu X, Zhou H, Guo H, Zhao X. Astaxanthin protects against renal fibrosis through inhibiting myofibroblast activation and promoting CD8+ T cell recruitment. Biochim Biophys Acta Gen Subj 2019; 1863:1360-1370. [DOI: 10.1016/j.bbagen.2019.05.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/19/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
|
17
|
Yip HK, Chen KH, Dubey NK, Sun CK, Deng YH, Su CW, Lo WC, Cheng HC, Deng WP. Cerebro- and renoprotective activities through platelet-derived biomaterials against cerebrorenal syndrome in rat model. Biomaterials 2019; 214:119227. [DOI: 10.1016/j.biomaterials.2019.119227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/18/2022]
|
18
|
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: 1.6] [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
|
19
|
Boldyreva MA, Shevchenko EK, Molokotina YD, Makarevich PI, Beloglazova IB, Zubkova ES, Dergilev KV, Tsokolaeva ZI, Penkov D, Hsu MN, Hu YC, Parfyonova YV. Transplantation of Adipose Stromal Cell Sheet Producing Hepatocyte Growth Factor Induces Pleiotropic Effect in Ischemic Skeletal Muscle. Int J Mol Sci 2019; 20:E3088. [PMID: 31238604 PMCID: PMC6627773 DOI: 10.3390/ijms20123088] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 01/08/2023] Open
Abstract
Cell therapy remains a promising approach for the treatment of cardiovascular diseases. In this regard, the contemporary trend is the development of methods to overcome low cell viability and enhance their regenerative potential. In the present study, we evaluated the therapeutic potential of gene-modified adipose-derived stromal cells (ADSC) that overexpress hepatocyte growth factor (HGF) in a mice hind limb ischemia model. Angiogenic and neuroprotective effects were assessed following ADSC transplantation in suspension or in the form of cell sheet. We found superior blood flow restoration, tissue vascularization and innervation, and fibrosis reduction after transplantation of HGF-producing ADSC sheet compared to other groups. We suggest that the observed effects are determined by pleiotropic effects of HGF, along with the multifactorial paracrine action of ADSC which remain viable and functionally active within the engineered cell construct. Thus, we demonstrated the high therapeutic potential of the utilized approach for skeletal muscle recovery after ischemic damage associated with complex tissue degenerative effects.
Collapse
MESH Headings
- Adipose Tissue/cytology
- Animals
- Cell Culture Techniques
- Cell Differentiation/genetics
- Cell Movement/drug effects
- Culture Media, Conditioned/pharmacology
- Disease Models, Animal
- Gene Expression
- Hepatocyte Growth Factor/biosynthesis
- Hepatocyte Growth Factor/genetics
- Humans
- Ischemia
- Mice
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/genetics
- Neuroglia/cytology
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neuronal Outgrowth/drug effects
- Stromal Cells/metabolism
- Stromal Cells/transplantation
Collapse
Affiliation(s)
- Maria A Boldyreva
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Evgeny K Shevchenko
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Yuliya D Molokotina
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
| | - Pavel I Makarevich
- Institute for Regenerative Medicine, Lomonosov Moscow State University, 119191 Moscow, Russia.
| | - Irina B Beloglazova
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Ekaterina S Zubkova
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Konstantin V Dergilev
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
| | - Zoya I Tsokolaeva
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Dmitry Penkov
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
| | - Mu-Nung Hsu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan.
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan.
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan.
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan.
| | - Yelena V Parfyonova
- National Medical Research Center of Cardiology, Russian Ministry of Health, 121552 Moscow, Russia.
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia.
| |
Collapse
|
20
|
Tubular cell and keratinocyte single-cell transcriptomics applied to lupus nephritis reveal type I IFN and fibrosis relevant pathways. Nat Immunol 2019; 20:915-927. [PMID: 31110316 PMCID: PMC6584054 DOI: 10.1038/s41590-019-0386-1] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 03/26/2019] [Indexed: 12/22/2022]
Abstract
The molecular and cellular processes that lead to renal damage and to the heterogeneity of lupus nephritis (LN) are not well understood. We applied single-cell RNA sequencing (scRNA-seq) to renal biopsies from patients with LN and evaluated skin biopsies as a potential source of diagnostic and prognostic markers of renal disease. Type I interferon (IFN) response signatures in tubular cells and in keratinocytes distinguished patients with LN from healthy control subjects. Moreover, a high IFN response signature and fibrotic signature in tubular cells were each associated with failure to respond to treatment. Analysis of tubular cells from patients with proliferative, membranous, and mixed LN indicated pathways relevant to inflammation and fibrosis, which offer insight into their histological differences. In summary, we applied scRNA-seq to LN to deconstruct its heterogeneity and identify novel targets for personalized approaches to therapy.
Collapse
|
21
|
Transforming growth factor β (TGFβ) and related molecules in chronic kidney disease (CKD). Clin Sci (Lond) 2019; 133:287-313. [DOI: 10.1042/cs20180438] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/04/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
|
22
|
Oka M, Sekiya S, Sakiyama R, Shimizu T, Nitta K. Hepatocyte Growth Factor-Secreting Mesothelial Cell Sheets Suppress Progressive Fibrosis in a Rat Model of CKD. J Am Soc Nephrol 2019; 30:261-276. [PMID: 30635373 DOI: 10.1681/asn.2018050556] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 11/24/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Although hepatocyte growth factor (HGF) has antifibrotic effects and is involved in angiogenesis and vasodilation, systemic administration of HGF to prevent kidney fibrosis is not a feasible strategy for suppressing interstitial fibrosis in patients with CKD. METHODS We investigated a novel therapy involving HGF transgenic cell sheets grown in culture from human mesothelial cells and administered to rats with unilateral ureteral obstruction (UUO). We compared progression of fibrosis in rats with UUO that received one of five interventions: HGF-transgenic mesothelial cell sheets transplanted to the kidney surface, HGF-transgenic mesothelial cell sheets transplanted to thigh, mesotherial cell sheets transplanted to kidney, no sheets, or HGF injections. RESULTS HGF transgenic cell sheets transplanted to the kidney strongly suppressed the induction of myofibroblasts and collagen in the kidney for 28 days; other interventions did not. Additionally, the HGF-secreting cell sheets ameliorated loss of peritubular capillaries and maintained renal blood flow. CONCLUSIONS These findings suggest that cell sheet therapy is a novel and promising strategy for inhibiting progressive fibrosis in CKD.
Collapse
Affiliation(s)
- Masatoshi Oka
- Department of Medicine, Kidney Center.,Institute of Advanced Biomedical Engineering and Science, and
| | - Sachiko Sekiya
- Institute of Advanced Biomedical Engineering and Science, and
| | - Ryoichi Sakiyama
- Department of Clinical Engineering, Tokyo Women's Medical University, Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, and
| | | |
Collapse
|
23
|
Miyagi H, Jalilian I, Murphy CJ, Thomasy SM. Modulation of human corneal stromal cell differentiation by hepatocyte growth factor and substratum compliance. Exp Eye Res 2018; 176:235-242. [PMID: 30193807 DOI: 10.1016/j.exer.2018.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 11/29/2022]
Abstract
Corneal wound healing is a complex process that consists of cellular integration of multiple soluble biochemical cues and cellular responses to biophysical attributes associated with the matrix of the wound space. Upon corneal stromal wounding, the transformation of corneal fibroblasts to myofibroblasts is promoted by transforming growth factor-β (TGFβ). This process is critical for wound healing; however, excessive persistence of myofibroblasts in the wound space has been associated with corneal fibrosis resulting in severe vision loss. The objective of this study was to determine the effect of hepatocyte growth factor (HGF), which can modulate TGFβ signaling, on corneal myofibroblast transformation by analyzing the expression of α-smooth muscle actin (αSMA) as a marker of myofibroblast phenotype particularly as it relates to biomechanical cues. Human corneal fibroblasts were cultured on tissue culture plastic (>1 GPa) or hydrogel substrates mimicking human normal or wounded corneal stiffness (25 and 75 kPa) in media containing TGFβ1 ± HGF. The expression of αSMA was analyzed by quantitative PCR, Western blot and immunocytochemistry. Cellular stiffness, which is correlated with cellular phenotype, was measured by atomic force microscopy (AFM). In primary human corneal fibroblasts, the mRNA expression of αSMA showed a clear dose response to TGFβ1. The expression was significantly suppressed when cells were incubated with 20 ng/ml HGF in the presence of 2 ng/ml of TGFβ1. The protein expression of αSMA induced by 5 ng/ml TGFβ1 was also decreased by 20 ng/ml of HGF. Cells cultured on hydrogels mimicking human normal (25 kPa) and fibrotic (75 kPa) cornea also showed an inhibitory effect of HGF on αSMA expression in the presence or absence of TGFβ1. Cellular stiffness was decreased by HGF in the presence of TGFβ1 as measured by AFM. In this study, we have demonstrated that HGF can suppress the myofibroblast phenotype promoted by TGFβ1 in human corneal stromal cells. These data suggest that HGF holds the potential as a therapeutic agent to improve wound healing outcomes by minimizing corneal fibrosis.
Collapse
Affiliation(s)
- Hidetaka Miyagi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA; Department of Ophthalmology and Visual Sciences, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Iman Jalilian
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Christopher J Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA; Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, CA, USA.
| | - Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA; Department of Ophthalmology & Vision Science, School of Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
24
|
Soluble cMet levels in urine are a significant prognostic biomarker for diabetic nephropathy. Sci Rep 2018; 8:12738. [PMID: 30143691 PMCID: PMC6109090 DOI: 10.1038/s41598-018-31121-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/13/2018] [Indexed: 12/23/2022] Open
Abstract
Hepatocyte growth factor and its receptor cMet activate biological pathways necessary for repair and regeneration following kidney injury. Here, we evaluated the clinical role of urinary cMet as a prognostic biomarker in diabetic nephropathy (DN). A total of 218 patients with DN were enrolled in this study. We examined the association of urine cMet levels and long-term outcomes in patients with DN. The levels of urinary cMet were higher in patients with decreased renal function than in patients with relatively preserved renal function (5.25 ± 9.62 ng/ml versus 1.86 ± 4.77 ng/ml, P = 0.001). A fully adjusted model revealed that a urinary cMet cutoff of 2.9 ng/mL was associated with a hazard ratio for end-stage renal disease of 2.33 (95% confidence interval 1.19–4.57, P = 0.014). The addition of urinary cMet to serum creatinine and proteinuria provided the highest net reclassification improvement. We found that in primary cultured human glomerular endothelial cells, TGFβ treatment induced fibrosis, and the protein expression levels of collagen I, collagen IV, fibronectin, and αSMA were decreased after administration of an agonistic cMet antibody. In conclusion, elevated levels of urinary cMet at the time of initial diagnosis could predict renal outcomes in patients with DN.
Collapse
|
25
|
Li YK, Ma DX, Wang ZM, Hu XF, Li SL, Tian HZ, Wang MJ, Shu YW, Yang J. The glucagon-like peptide-1 (GLP-1) analog liraglutide attenuates renal fibrosis. Pharmacol Res 2018. [DOI: 10.1016/j.phrs.2018.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
26
|
Abstract
PURPOSE OF REVIEW Multiple experimental and clinical studies have identified pathways by which uric acid may facilitate the development and progression of chronic kidney disease (CKD) in people with diabetes. However, it remains uncertain if the association of uric acid with CKD represents a pathogenic effect or merely reflects renal impairment. RECENT FINDINGS In contrast to many published reports, a recent Mendelian randomization study did not identify a causal link between uric acid and CKD in people with type 1 diabetes. Two recent multicenter randomized control trials, Preventing Early Renal Function Loss in Diabetes (PERL) and FEbuxostat versus placebo rAndomized controlled Trial regarding reduced renal function in patients with Hyperuricemia complicated by chRonic kidney disease stage 3 (FEATHER), were recently designed to assess if uric acid lowering slows progression of CKD. We review the evidence supporting a role for uric acid in the pathogenesis of CKD in people with diabetes and the putative benefits of uric acid lowering.
Collapse
Affiliation(s)
- Ambreen Gul
- Dialysis Clinic, Inc., Quality Management, 1500 Indian School Rd. NE, Albuquerque, NM, 87102, USA
| | - Philip Zager
- Dialysis Clinic, Inc., Quality Management, 1500 Indian School Rd. NE, Albuquerque, NM, 87102, USA.
- University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
27
|
Cai T, Sun D, Duan Y, Qiu Y, Dai C, Yang J, He W. FHL2 promotes tubular epithelial-to-mesenchymal transition through modulating β-catenin signalling. J Cell Mol Med 2017; 22:1684-1695. [PMID: 29193729 PMCID: PMC5824423 DOI: 10.1111/jcmm.13446] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022] Open
Abstract
β-Catenin signalling plays an important role in regulating tubular epithelial-to-mesenchymal transition (EMT), an indispensable programme for driving renal fibrosis. As an adapter protein, four and a half LIM domain protein 2 (FHL2) acts as a coregulator of β-catenin in several other cell types. To determine whether FHL2 affects β-catenin signalling and thus is involved in tubular EMT, we examined its expression and function in the process of TGF-β1-induced EMT. FHL2 mRNA and protein were induced by TGF-β1 in rat tubular epithelial cells (NRK-52E), an effect that intracellular Smad signalling was required. Ectopic expression of FHL2 inhibited E-cadherin and enhanced α-smooth muscle actin (α-SMA) and fibronectin expression, whereas knockdown of FHL2 partially restored E-cadherin and reduced α-SMA and fibronectin induction stimulated by TGF-β1. Overexpression of FHL2 increased β-catenin dephosphorylation (Ser37/Thr41), nuclear translocation and β-catenin-mediated transcription and up-regulated expression of β-catenin target, EMT-related genes, such as Snail, Twist, vimentin, plasminogen activator inhibitor-1 and matrix metalloproteinase-7. Conversely, knockdown of FHL2 increased β-catenin phosphorylation (Ser33/37/Thr41), decreased its nuclear translocation and inhibited β-catenin-mediated transcription and target genes expression. TGF-β1 induced a FHL2/β-catenin interaction in NRK-52E cells, especially in the nuclei. In a mouse model of obstructive nephropathy, FHL2 mRNA and protein were induced in a time-dependent fashion, and the extent and pattern of renal β-catenin activation were positively correlated with FHL2 induction. Collectively, this study suggests that FHL2, via modulating β-catenin signalling, may implicate in regulation of TGF-β1-mediated tubular EMT and could be a potential therapeutic target for fibrotic kidney disease.
Collapse
Affiliation(s)
- Ting Cai
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danqin Sun
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Duan
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yumei Qiu
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunsun Dai
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weichun He
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
28
|
Cannito S, Novo E, Parola M. Therapeutic pro-fibrogenic signaling pathways in fibroblasts. Adv Drug Deliv Rev 2017; 121:57-84. [PMID: 28578015 DOI: 10.1016/j.addr.2017.05.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/28/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
Myofibroblasts (MFs) play a critical role in the progression of chronic inflammatory and fibroproliferative diseases in different tissues/organs, whatever the etiology. Fibrosis is preceded and sustained by persistent injury and inflammatory response in a profibrogenic scenario involving mutual interactions, operated by several mediators and pathways, of MFs and related precursor cells with innate immunity cells and virtually any cell type in a defined tissue. These interactions, mediators and related signaling pathways are critical in initiating and perpetuating the differentiation of precursor cells into MFs that in different tissues share peculiar traits and phenotypic responses, including the ability to proliferate, produce ECM components, migrate and contribute to the modulation of inflammatory response and tissue angiogenesis. Literature studies related to liver, lung and kidney fibrosis have outlined a number of MF-related core regulatory fibrogenic signaling pathways conserved across these different organs and potentially targetable in order to develop effective antifibrotic therapeutic strategies.
Collapse
|
29
|
Wang LY, Diao ZL, Zheng JF, Wu YR, Zhang QD, Liu WH. Apelin attenuates TGF-β1-induced epithelial to mesenchymal transition via activation of PKC-ε in human renal tubular epithelial cells. Peptides 2017; 96:44-52. [PMID: 28847490 DOI: 10.1016/j.peptides.2017.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 12/13/2022]
Abstract
Epithelial to mesenchymal transition (EMT), a process whereby fully differentiated epithelial cells transition to a mesenchymal phenotype, has been implicated in the pathogenesis of renal fibrosis. Apelin, a bioactive peptide, has recently been recognized to protect against renal profibrotic activity, but the underlying mechanism has not yet been elucidated. In this study, we investigated the regulation of EMT in the presence of apelin-13 in vitro. Expression of the mesenchymal marker alpha-smooth muscle actin (α-SMA) and the epithelial marker E-cadherin was examined by immunofluorescence and western blotting in transforming growth factor beta 1 (TGF-β1)-stimulated human proximal tubular epithelial cells. Expression of extracellular matrix, fibronectin and collagen-I was examined by quantitative real-time PCR and ELISA. F13A, an antagonist of the apelin receptor APJ, and small interfering RNA targeting protein kinase C epsilon (PKC-ε) were used to explore the relevant signaling pathways. Apelin attenuated TGF-β1-induced EMT, and inhibited the EMT-associated increase in α-SMA, loss of E-cadherin, and secretion of extracellular matrix. Moreover, apelin activated PKC-ε in tubular epithelial cells, which in turn decreased phospho-Smad2/3 levels and increased Smad-7 levels. APJ inhibition or PKC-ε deletion diminished apelin-induced modulation of Smad signaling and suppression of tubular EMT. Our findings identify a novel PKC-ε-dependent mechanism in which apelin suppresses TGF-β1-mediated activation of Smad signaling pathways and thereby inhibits tubular EMT. These results suggest that apelin may be a new agent that can suppress renal fibrosis and retard chronic kidney disease progression.
Collapse
Affiliation(s)
- Li-Yan Wang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zong-Li Diao
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jun-Fang Zheng
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China
| | - Yi-Ru Wu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Qi-Dong Zhang
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wen-Hu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
30
|
Matheus LHG, Simão GM, Amaral TA, Brito RBO, Malta CS, Matos YST, Santana AC, Rodrigues GGC, Albejante MC, Bach EE, Dalboni MA, Camacho CP, Dellê H. Indoleamine 2, 3-dioxygenase (IDO) increases during renal fibrogenesis and its inhibition potentiates TGF-β 1-induced epithelial to mesenchymal transition. BMC Nephrol 2017; 18:287. [PMID: 28877670 PMCID: PMC5585959 DOI: 10.1186/s12882-017-0702-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/22/2017] [Indexed: 01/01/2023] Open
Abstract
Background Indoleamine 2, 3-dioxygenase (IDO) is an immunomodulatory molecule that has been implicated in several biological processes. Although IDO has been linked with some renal diseases, its role in renal fibrosis is still unclear. Because IDO may be modulated by TGF-β1, a potent fibrogenic molecule, we hypothesized that IDO could be involved in renal fibrosis, especially acting in the TGF-β1-induced tubular epithelial-mesenchymal transition (EMT). We analyzed the IDO expression and activity in a model of renal fibrogenesis, and the effect of the IDO inhibitor 1-methyl-tryptophan (MT) on TGF-β1-induced EMT using tubular cell culture. Methods Male Wistar rats where submited to 7 days of UUO. Non-obstructed kidneys (CL) and kidneys from SHAM rats were used as controls. Masson’s Tricrome and macrophages counting were used to chatacterize the tissue fibrosis. The EMT was analysed though immunohistochemistry and qRT-PCR. Immunohistochemestry in tissue has used to show IDO expression. MDCK cells were incubated with TGF- β1 to analyse IDO expression. Additionally, effects of TGF- β1 and the inhibition of IDO over the EMT process was acessed by immunoessays and scrath wound essay. Results IDO was markedly expressed in cortical and medular tubules of the UUO kidneys. Similarly to the immunolocalizaton of TGF- β1, accompanied by loss of e-cadherin expression and an increase of mesenchymal markers. Results in vitro with MDCK cells, showed that IDO was increased after stimulus with TGF-β1, and treatment with MT potentiated its expression. MDCK stimulated with TGF-β1 had higher migratory activity (scratch-wound assay), which was exacerbated by MT treatment. Conclusions IDO is constitutively expressed in tubular cells and increases during renal fibrogenesis. Although IDO is induced by TGF-β1 in tubular cells, its chemical inhibitor acts as a profibrotic agent.
Collapse
Affiliation(s)
- Luiz Henrique Gomes Matheus
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Gislene Mendes Simão
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Taíssa Altieri Amaral
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Rodrigo Barbosa Oliveira Brito
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Camila Soares Malta
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Yves Silva Teles Matos
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Alexandre Chagas Santana
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Gabriela Gomes Cardoso Rodrigues
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Maria Clara Albejante
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Erna Elisabeth Bach
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Maria Aparecida Dalboni
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Cleber Pinto Camacho
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Humberto Dellê
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil.
| |
Collapse
|
31
|
Zhang CY, Zhu JY, Ye Y, Zhang M, Zhang LJ, Wang SJ, Song YN, Zhang H. Erhuang Formula ameliorates renal damage in adenine-induced chronic renal failure rats via inhibiting inflammatory and fibrotic responses. Biomed Pharmacother 2017; 95:520-528. [PMID: 28866419 DOI: 10.1016/j.biopha.2017.08.115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/21/2022] Open
Abstract
AIMS The present study aimed to evaluate the protective effects of Erhuang Formula (EHF) and explore its pharmacological mechanisms on adenine-induced chronic renal failure (CRF). MATERIALS AND METHODS The compounds in EHF were analyzed by HPLC/MS. Adenine-induced CRF rats were administrated by EHF. The effects were evaluated by renal function examination and histology staining. Immunostaining of some proteins related cell adhesion was performedin renal tissues, including E-cadherin, β-catenin, fibronectin and laminin. The qRT-PCR was carried out determination of gene expression related inflammation and fibrosis including NF-κB, TNF-α, TGF-β1, α-SMA and osteopontin (OPN). RESULTS Ten compounds in EHF were identified including liquiritigenin, farnesene, vaccarin, pachymic acid, cycloastragenol, astilbin, 3,5,6,7,8,3',4'-heptemthoxyflavone, physcion, emodin and curzerene. Abnormal renal function and histology had significant improvements by EHF treatment. The protein expression of β-catenin, fibronectin and laminin were significantly increased and the protein expression of E-cadherin significantly decreased in CRF groups. However, these protein expressions were restored to normal levels in EHF group. Furthermore, low expression of PPARγ and high expression of NF-κB, TNF-α, TGF-β1, α-SMA and OPN were substantially restored by EHF treatment in a dose-dependent manner. CONCLUSIONS EHF ameliorated renal damage in adenine-induced CRF rats, and the mechanisms might involve in the inhibition of inflammatory and fibrotic responses and the regulation of PPARγ, NF-κB and TGF-β signaling pathways.
Collapse
Affiliation(s)
- Chun-Yan Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of TCM, 358 Datong Road, Pudong, Shanghai, 200137, China
| | - Jian-Yong Zhu
- Central Laboratory, Seventh People's Hospital of Shanghai University of TCM, 358 Datong Road, Pudong, Shanghai, 200137, China
| | - Ying Ye
- Central Laboratory, Seventh People's Hospital of Shanghai University of TCM, 358 Datong Road, Pudong, Shanghai, 200137, China
| | - Miao Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of TCM, 358 Datong Road, Pudong, Shanghai, 200137, China
| | - Li-Jun Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of TCM, 358 Datong Road, Pudong, Shanghai, 200137, China
| | - Su-Juan Wang
- Central Laboratory, Seventh People's Hospital of Shanghai University of TCM, 358 Datong Road, Pudong, Shanghai, 200137, China
| | - Ya-Nan Song
- Central Laboratory, Seventh People's Hospital of Shanghai University of TCM, 358 Datong Road, Pudong, Shanghai, 200137, China.
| | - Hong Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of TCM, 358 Datong Road, Pudong, Shanghai, 200137, China.
| |
Collapse
|
32
|
Simvastatin treatment boosts benefits of apoptotic cell infusion in murine lung fibrosis. Cell Death Dis 2017; 8:e2860. [PMID: 28594406 PMCID: PMC5520916 DOI: 10.1038/cddis.2017.260] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 05/08/2017] [Accepted: 05/08/2017] [Indexed: 01/11/2023]
Abstract
A single early-phase infusion of apoptotic cells can inhibit bleomycin-induced lung inflammation and fibrosis; however, it is unknown whether these effects can be enhanced with additional infusions and/or statin treatment. Here, we investigated whether an increased frequency of apoptotic cell injection, with or without efferocytosis enhancer simvastatin, facilitates therapeutic efficacy. An additional injection of apoptotic cells during the intermediate phase (7 days post-bleomycin treatment) or simvastatin administration alone on days 7–13 post-treatment did not promote anti-fibrotic responses beyond those induced by a single early apoptotic cell infusion alone. Additional administration of apoptotic cells with simvastatin further enhanced the efferocytic ability of alveolar macrophages and PPARγ activity, and induced hepatocyte growth factor and interleukin-10 expression, in alveolar macrophages and lung tissue. Additional administration of apoptotic cells with simvastatin also reduced mRNA expression of bleomycin-induced epithelial-mesenchymal transition (EMT) markers in isolated alveolar type II epithelial cells, fibrotic markers in fibroblasts, and hydroxyproline in lung tissue. Enhanced anti-EMT and anti-fibrotic efficacy was confirmed by immunofluorescence and trichrome staining of lung tissue. This suggests that additional administration of apoptotic cells with simvastatin during the intermediate phase of bleomycin-induced lung fibrosis may boost the anti-fibrotic properties of early apoptotic cell infusion.
Collapse
|
33
|
Zhao Y, Yin Z, Li H, Fan J, Yang S, Chen C, Wang DW. MiR-30c protects diabetic nephropathy by suppressing epithelial-to-mesenchymal transition in db/db mice. Aging Cell 2017; 16:387-400. [PMID: 28127848 PMCID: PMC5334541 DOI: 10.1111/acel.12563] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2016] [Indexed: 12/24/2022] Open
Abstract
Epithelial‐to‐mesenchymal transition (EMT) plays a significant role in tubulointerstitial fibrosis, which is a hallmark of diabetic nephropathy. Thus, identifying the mechanisms of EMT activation could be meaningful. In this study, loss of miR‐30c accompanied with increased EMT was observed in renal tubules of db/db mice and cultured HK2 cells exposed to high glucose. To further explore the roles of miR‐30c in EMT and tubulointerstitial fibrosis, recombinant adeno‐associated viral vector was applied to manipulate the expression of miR‐30c. In vivo study showed that overexpression of miR‐30c suppressed EMT, attenuated renal tubulointerstitial fibrosis and reduced proteinuria, serum creatinine, and BUN levels. In addition, Snail1 was identified as a direct target of miR‐30c by Ago2 co‐immunoprecipitation, luciferase reporter, and Western blot assays. Downregulating Snail1 by siRNA reduced high glucose‐induced EMT in HK2 cells, and miR‐30c mimicked the effects. Moreover, miR‐30c inhibited Snail1‐TGF‐β1 axis in tubular epithelial cells undergoing EMT and thereby impeded the release of TGF‐β1; oppositely, knockdown of miR‐30c enhanced the secretion of TGF‐β1 from epitheliums and significantly promoted proliferation of fibroblasts and fibrogenesis of myofibroblasts, aggravated tubulointerstitial fibrosis, and dysfunction of diabetic nephropathy. These results suggest a protective role of miR‐30c against diabetic nephropathy by suppressing EMT via inhibiting Snail1‐TGF‐β1 pathway.
Collapse
Affiliation(s)
- Yanru Zhao
- Division of Cardiology; Department of Internal Medicine; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan 430030 China
| | - Zhongwei Yin
- Division of Cardiology; Department of Internal Medicine; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan 430030 China
| | - Huaping Li
- Division of Cardiology; Department of Internal Medicine; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan 430030 China
| | - Jiahui Fan
- Division of Cardiology; Department of Internal Medicine; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan 430030 China
| | - Shenglan Yang
- Department of Cardiology; The First Affiliated Hospital of Chongqing Medical University; Chongqing 400042 China
| | - Chen Chen
- Division of Cardiology; Department of Internal Medicine; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan 430030 China
| | - Dao Wen Wang
- Division of Cardiology; Department of Internal Medicine; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan 430030 China
| |
Collapse
|
34
|
Moritz RJ, LeBaron RG, Phelix CF, Rupaimoole R, Kim HS, Tsin A, Asmis R. Macrophage TGF- β1 and the Proapoptotic Extracellular Matrix Protein BIGH3 Induce Renal Cell Apoptosis in Prediabetic and Diabetic Conditions. ACTA ACUST UNITED AC 2017; 7:496-510. [PMID: 28149671 PMCID: PMC5279341 DOI: 10.4236/ijcm.2016.77055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Metabolically stressed kidney is in part characterized by infiltrating macrophages and macrophage-derived TGF-β1 that promote the synthesis of various ECM molecules. TGF-β1 strongly enhances the expression of the gene TGFBI that encodes a cell-adhesion class, proapoptotic ECM protein called BIGH3. We hypothesized that in a diabetic environment a relationship between infiltrating macrophages, macrophage-derived TGF-β1, and BIGH3 protein promotes renal cell death. To investigate this hypothesis, we used our mouse model of diabetic complications. Mice on a high-fat diet developed hypercholesterolemia, and exposure to streptozotocin rendered hypercholesterolemic mice diabetic. Immunohistochemical images show increased macrophage infiltration and BIGH3 protein in the kidney cortices of hypercholesterolemic and diabetic mice. Macrophages induced a two-fold increase in BIGH3 expression and an 86% increase in renal proximal tubule epithelial cell apoptosis. TGF-β1 antibody and TGF-β1 receptor chemical antagonist blocked macrophage-induced apoptosis. BIGH3 antibody completely blocked apoptosis that was induced by TGF-β1, and blocked apoptosis induced by exogenous recombinant BIGH3. These results uncover a distinctive interplay of macrophage-derived TGF-β1, BIGH3 protein, and apoptosis, and indicate that BIGH3 is central in a novel pathway that promotes diabetic nephropathy. Macrophage TGF-β1 and BIGH3 are identified as prediabetic biomarkers, and potential therapeutic targets for intervention in prediabetic and diabetic individuals.
Collapse
Affiliation(s)
- Robert J Moritz
- Department of Biology, University of Texas at San Antonio, San Antonio, USA
| | - Richard G LeBaron
- Department of Biology, University of Texas at San Antonio, San Antonio, USA
| | - Clyde F Phelix
- Department of Biology, University of Texas at San Antonio, San Antonio, USA
| | - Rajesha Rupaimoole
- Department of Biology, University of Texas at San Antonio, San Antonio, USA
| | - Hong Seok Kim
- Departments of Biochemistry and Clinical Laboratory Sciences, School of Health Professions, University of Texas Health Science Center at San Antonio, San Antonio, USA
| | - Andrew Tsin
- Department of Biology, University of Texas at San Antonio, San Antonio, USA
| | - Reto Asmis
- Departments of Biochemistry and Clinical Laboratory Sciences, School of Health Professions, University of Texas Health Science Center at San Antonio, San Antonio, USA
| |
Collapse
|
35
|
Moghadam A, Khozani TT, Mafi A, Namavar MR, Dehghani F. Effects of Platelet-Rich Plasma on Kidney Regeneration in Gentamicin-Induced Nephrotoxicity. J Korean Med Sci 2017; 32:13-21. [PMID: 27914126 PMCID: PMC5143284 DOI: 10.3346/jkms.2017.32.1.13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/20/2016] [Indexed: 12/25/2022] Open
Abstract
Platelet-rich plasma (PRP) as a source of growth factors may induce tissue repairing and improve fibrosis. This study aimed to assess the effects of PRP on kidney regeneration and fibrosis in gentamicin (GM)-induced nephrotoxicity rat model by stereological study. Thirty-two male rats were selected. Nephrotoxicity was induced in animals by administration of GM (80 mg/kg/daily, intraperitoneally [IP], 8 day) and animals were treated by PRP (100 μL, intra-cortical injection using surgical microscopy, single dose). Blood samples were collected for determine blood urea nitrogen (BUN) and creatinine (Cr) before and after PRP therapy. At the end of experiment, right kidneys were sectioned by Isotropic Uniform Random (IUR) method and stained with H & E and Masson's Trichrome. The stereological methods were used for estimating the changes in different structures of kidney. PRP increased the number of epithelial cells in convoluted tubules, and decreased the volume of connective tissue, renal corpuscles and glomeruli in GM-treated animals (P < 0.05). Our findings indicate that PRP had beneficial effects on proliferation of epithelial cells in convoluted tubules and ameliorated GM-induced fibrosis.
Collapse
Affiliation(s)
- Abbas Moghadam
- Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Afsaneh Mafi
- Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Namavar
- Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Dehghani
- Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
36
|
Feng J, Ge S, Zhang L, Che H, Liang C. Aortic dissection is associated with reduced polycystin-1 expression, an abnormality that leads to increased ERK phosphorylation in vascular smooth muscle cells. Eur J Histochem 2016; 60:2711. [PMID: 28076932 PMCID: PMC5381529 DOI: 10.4081/ejh.2016.2711] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/21/2016] [Accepted: 11/21/2016] [Indexed: 12/17/2022] Open
Abstract
The vascular smooth muscle cell (VSMC) phenotypic switch is a key pathophysiological change in various cardiovascular diseases, such as aortic dissection (AD), with a high morbidity. Polycystin-1 (PC1) is significantly downregulated in the VSMCs of AD patients. PC1 is an integral membrane glycoprotein and kinase that regulates different biological processes, including cell proliferation, apoptosis, and cell polarity. However, the role of PC1 in intracellular signaling pathways remains poorly understood. In this study, PC1 downregulation in VSMCs promoted the expression of SM22α, ACTA2 and calponin 1 (CNN1) proteins. Furthermore, PC1 downregulation in VSMCs upregulated phospho-MEK, phospho-ERK and myc, but did not change phospho-JNK and phospho-p38. These findings suggest that the MEK/ERK/myc signaling pathway is involved in PC1-mediated human VSMC phenotypic switch. Opposite results were observed when an ERK inhibitor was used in VSMCs downregulated by PC1. When the C-terminal domain of PC1 (PC1 C-tail) was overexpressed in VSMCs, the expression levels of phosphor-ERK, myc, SM22α, ACTA2 and CNN1 proteins were downregulated. The group with the overexpressed mutant protein (S4166A) in the PC1 C-tail showed similar results to the group with the downregulated PC1 in VSMCs. These results suggest that the Ser at the 4166 site in PC1 is crucial in the PC1 mediated MEK/ERK/myc signaling pathway, which might be the key pathophysiological cause of AD.
Collapse
Affiliation(s)
- J Feng
- The First Affiliated Hospital of Anhui Medical University, Department of Cardiovascular Surgery.
| | | | | | | | | |
Collapse
|
37
|
The cellular and signalling alterations conducted by TGF-β contributing to renal fibrosis. Cytokine 2016; 88:115-125. [DOI: 10.1016/j.cyto.2016.08.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 01/08/2023]
|
38
|
Huang H, Zheng F, Dong X, Wu F, Wu T, Li H. Allicin inhibits tubular epithelial-myofibroblast transdifferentiation under high glucose conditions in vitro. Exp Ther Med 2016; 13:254-262. [PMID: 28123498 DOI: 10.3892/etm.2016.3913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/04/2016] [Indexed: 12/16/2022] Open
Abstract
Previous studies have suggested that tubular epithelial-mesenchymal transition (EMT) is an important event in renal tubulointerstitial fibrosis, which is a clinical characteristic of diabetic nephropathy. The present study aimed to investigate the effect of allicin, the major biological active component of garlic, on the EMT of a human renal proximal tubular epithelial cell line (HK-2) cultured under high glucose concentrations. HK-2 cells were exposed for 48 h to 5.5 or 25 mmol/l D-glucose, 25 mmol/l D-glucose plus allicin (2.5, 5, 10 or 20 µg/ml) or 25 mmol/l D-glucose plus 20 µmol/l PD98059, a selective inhibitor of the mitogen activated protein kinase/extracellular signal-regulated kinase (ERK) signaling pathway. The EMT of HK-2 cells was assessed by analyzing the protein expression of E-cadherin, α-smooth muscle actin (α-SMA), vimentin and collagen I via immunocytochemistry. In addition, reverse transcription-quantitative polymerase chain reaction and western blotting were used to detect the expression levels of transforming growth factor (TGF)-β1 and phosphorylated (p)-ERK1/2. Marked morphological changes were observed in HK-2 cells cultured under high glucose conditions, and these changes were abrogated by simultaneous incubation with allicin and PD98059. The expression levels of α-SMA, vimentin and collagen I were significantly increased in HK-2 cells cultured under high glucose conditions, as compared with those cultured under normal glucose conditions (P<0.01). Conversely, the expression levels of E-cadherin were significantly decreased upon stimulation with high glucose (P<0.01). Furthermore, the expression levels of TGF-β1 and p-ERK1/2 were significantly upregulated in HK-2 cells cultured under high glucose conditions, as compared with those cultured under normal glucose conditions (P<0.05). Allicin partially reversed the high-glucose-induced increase in α-SMA, vimentin and collagen I expression (P<0.01 at 20 µg/ml), increased the expression of E-cadherin, and significantly downregulated the high glucose-induced expression of TGF-β1 and p-ERK1/2 in a dose-dependent manner (P<0.05). The results of the present study suggested that high glucose concentrations induced the EMT of HK-2 cells, and that allicin was able to inhibit the EMT, potentially via regulation of the ERK1/2-TGF-β1 signaling pathway.
Collapse
Affiliation(s)
- Hong Huang
- Department of Endocrinology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Fenping Zheng
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Xuehong Dong
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Fang Wu
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Tianfeng Wu
- Department of Endocrinology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Hong Li
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
39
|
Cho JH, Ryu HM, Oh EJ, Yook JM, Ahn JS, Jung HY, Choi JY, Park SH, Kim YL, Kwak IS, Kim CD. Alpha1-Antitrypsin Attenuates Renal Fibrosis by Inhibiting TGF-β1-Induced Epithelial Mesenchymal Transition. PLoS One 2016; 11:e0162186. [PMID: 27607429 PMCID: PMC5015906 DOI: 10.1371/journal.pone.0162186] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/18/2016] [Indexed: 01/12/2023] Open
Abstract
Alpha1-antitrypsin (AAT) exerts its anti-inflammatory effect through regulating the activity of serine proteinases. This study evaluated the inhibitory effects of AAT against the transforming growth factor (TGF)-β1 induced epithelial-to-mesenchymal transition (EMT) in unilateral ureter obstruction (UUO) mice and Madin-Darby canine kidney (MDCK) cells. C57BL/6 mice with induced UUO were injected intraperitoneally with AAT (80 mg/Kg) or vehicle for 7 days. MDCK cells were treated with TGF-β1 (2 ng/mL) for 48 hours to induce EMT, and co-treated with AAT (10 mg/mL) to inhibit the EMT. Masson’s trichrome and Sirius red staining was used to estimate the extent of renal fibrosis in UUO mice. The expression of alpha-smooth muscle actin (α-SMA), vimentin, fibronectin, collagen I, and E-cadherin in MDCK cells and kidney tissue were evaluated. Masson’s and Sirius red staining revealed that the area of renal fibrosis was significantly smaller in AAT treated UUO group compared with that of UUO and vehicle treated UUO groups. AAT treatment attenuated upregulation of Smad2/3 phosphorylation in UUO mouse model. Co-treatment of MDCK cells with TGF-β1 and AAT significantly attenuated the changes in the expression of α-SMA, vimentin, fibronectin, collagen I, and E-cadherin. AAT also decreased the phosphorylated Smad3 expression and the phosphorylated Smad3/Smad3 ratio in MDCK cells. AAT treatment inhibited EMT induced by TGF-β1 in MDCK cells and attenuated renal fibrosis in the UUO mouse model. The results of this work suggest that AAT could inhibit the process of EMT through the suppression of TGF-β/Smad3 signaling.
Collapse
Affiliation(s)
- Jang-Hee Cho
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Hye-Myung Ryu
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Eun-Joo Oh
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Ju-Min Yook
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Ji-Sun Ahn
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Hee-Yeon Jung
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Ji-Young Choi
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Sun-Hee Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Yong-Lim Kim
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Ihm Soo Kwak
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
- * E-mail: (CDK); (ISK)
| | - Chan-Duck Kim
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
- * E-mail: (CDK); (ISK)
| |
Collapse
|
40
|
Tang H, Fan D, Lei CT, Ye C, Gao P, Chen S, Meng XF, Su H, Zhang C. MAD2B promotes tubular epithelial-to-mesenchymal transition and renal tubulointerstitial fibrosis via Skp2. J Mol Med (Berl) 2016; 94:1297-1307. [PMID: 27488450 DOI: 10.1007/s00109-016-1448-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/16/2016] [Accepted: 07/17/2016] [Indexed: 12/11/2022]
Abstract
The mitotic arrest deficient protein MAD2B is a well-defined anaphase-promoting complex/cyclosome (APC/C) inhibitor and a small subunit of DNA polymerase zeta. It is critical for mitotic control and DNA repair. However, the pathological role of MAD2B in kidney diseases has not been fully elucidated. In the present study, we aim to explore the role of MAD2B in the pathogenesis of renal tubulointerstitial fibrosis (TIF) and the underlying mechanism. By immunofluorescence and immunohistochemistry, we found an obvious MAD2B enhancement in tubular area of TIF patients and unilateral ureteral obstruction (UUO) mice. In vitro, transforming growth factor-β1 (TGF-β1) induced a time-dependent MAD2B accumulation prior to tubular epithelial-to-mesenchymal transition (EMT) in a rat proximal tubular epithelial cell line, NRK-52E. Knocking down MAD2B using siRNA dramatically inhibited TGF-β1-induced tubular EMT process and subsequent extracellular matrix (ECM) production. We also found that Skp2, a confirmed APC/C-CDH1 substrate and E-cadherin destroyer, was increased in TGF-β1-treated proximal tubular epithelial cells, which could be blocked by MAD2B depletion. In addition, Skp2 expression was also found to be increased in the renal tubular area of UUO mice. Locally knocking down MAD2B expression in the renal cortex using lentiviral transfection inhibited Skp2 expression, tubular EMT, and subsequent ECM accumulation. Taken together, our data suggests a pro-fibrotic role of MAD2B in the pathogenesis of tubular EMT and TIF by inducing Skp2 expression. MAD2B might be a potential target of promising interventions for renal TIF. KEY MESSAGES Renal fibrosis activates MAD2B expression in renal tubules of human and mouse. TGF-β1 contributes to MAD2B enhancement in rat tubular epithelial cells. MAD2B depletion alleviates renal tubulointerstitial fibrosis in vivo and in vitro. MAD2B promotes EMT transition in rat tubular epithelial cells by inducing Skp2.
Collapse
Affiliation(s)
- Hui Tang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Di Fan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun-Tao Lei
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chen Ye
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Pan Gao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shan Chen
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xian-Fang Meng
- Department of Neurobiology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
41
|
Singh S, Manson SR, Lee H, Kim Y, Liu T, Guo Q, Geminiani JJ, Austin PF, Chen YM. Tubular Overexpression of Angiopoietin-1 Attenuates Renal Fibrosis. PLoS One 2016; 11:e0158908. [PMID: 27454431 PMCID: PMC4959721 DOI: 10.1371/journal.pone.0158908] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/23/2016] [Indexed: 01/22/2023] Open
Abstract
Emerging evidence has highlighted the pivotal role of microvasculature injury in the development and progression of renal fibrosis. Angiopoietin-1 (Ang-1) is a secreted vascular growth factor that binds to the endothelial-specific Tie2 receptor. Ang-1/Tie2 signaling is critical for regulating blood vessel development and modulating vascular response after injury, but is dispensable in mature, quiescent vessels. Although dysregulation of vascular endothelial growth factor (VEGF) signaling has been well studied in renal pathologies, much less is known about the role of the Ang-1/Tie2 pathway in renal interstitial fibrosis. Previous studies have shown contradicting effects of overexpressing Ang-1 systemically on renal tubulointerstitial fibrosis when different engineered forms of Ang-1 are used. Here, we investigated the impact of site-directed expression of native Ang-1 on the renal fibrogenic process and peritubular capillary network by exploiting a conditional transgenic mouse system [Pax8-rtTA/(TetO)7 Ang-1] that allows increased tubular Ang-1 production in adult mice. Using a murine unilateral ureteral obstruction (UUO) fibrosis model, we demonstrate that targeted Ang-1 overexpression attenuates myofibroblast activation and interstitial collagen I accumulation, inhibits the upregulation of transforming growth factor β1 and subsequent phosphorylation of Smad 2/3, dampens renal inflammation, and stimulates the growth of peritubular capillaries in the obstructed kidney. Our results suggest that Ang-1 is a potential therapeutic agent for targeting microvasculature injury in renal fibrosis without compromising the physiologically normal vasculature in humans.
Collapse
Affiliation(s)
- Sudhir Singh
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Scott R. Manson
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Heedoo Lee
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Yeawon Kim
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Tuoen Liu
- Oncology Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Qiusha Guo
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Julio J. Geminiani
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Paul F. Austin
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Ying Maggie Chen
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
- * E-mail:
| |
Collapse
|
42
|
Zhang LM, Liu JH, Xue CB, Li MQ, Xing S, Zhang X, He WT, Jiang FC, Lu X, Zhou P. Pharmacological inhibition of MyD88 homodimerization counteracts renal ischemia reperfusion-induced progressive renal injury in vivo and in vitro. Sci Rep 2016; 6:26954. [PMID: 27246399 PMCID: PMC4887891 DOI: 10.1038/srep26954] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 05/11/2016] [Indexed: 12/19/2022] Open
Abstract
The activation of innate immunity via myeloid differentiation factor 88 (MyD88) contributes to ischemia reperfusion (I/R) induced acute kidney injury (AKI) and chronic kidney injury. However, since there have not yet been any effective therapy, the exact pharmacological role of MyD88 in the prevention and treatment of renal ischemia reperfusion injury (IRI) is not known. We designed a small molecular compound, TJ-M2010-2, which inhibited MyD88 homodimerization. We used an established unilateral I/R mouse model. All mice undergoing 80 min ischemia through uninephrectomy died within five days without intervention. However, treatment with TJ-M2010-2 alone significantly improved the survival rate to 58.3%. Co-treatment of TJ-M2010-2 with the CD154 antagonist increased survival rates up to 100%. Twenty-eight days post-I/R of 60 min ischemia without nephrectomy, TJ-M2010-2 markedly attenuated renal interstitial and inhibited TGF-β1-induced epithelial-mesenchymal transition (EMT) of renal tubular epithelial cells. Furthermore, TJ-M2010-2 remarkably inhibited TLR/MyD88 signaling in vivo and in vitro. In conclusion, our findings highlight the promising clinical potential of MyD88 inhibitor in preventing and treating acute or chronic renal I/R injuries, and the therapeutic functionality of dual-system inhibition strategy in IRI-induced AKI. Moreover, MyD88 inhibition ameliorates renal I/R injury-induced tubular interstitial fibrosis by suppressing EMT.
Collapse
Affiliation(s)
- Li-Min Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Health, and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan 430030, China
| | - Jian-Hua Liu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Health, and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan 430030, China
| | - Cheng-Biao Xue
- Institute of Hepatobiliary Diseases of Wuhan University, Zhongnan Hospital of Wuhan University; Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan 430071, China
| | | | - Shuai Xing
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Health, and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan 430030, China
| | - Xue Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Health, and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan 430030, China
| | - Wen-Tao He
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng-Chao Jiang
- Academy of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xia Lu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Health, and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan 430030, China
| | - Ping Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Health, and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan 430030, China
| |
Collapse
|
43
|
Hara S, Kawano M, Mizushima I, Harada K, Takata T, Saeki T, Ubara Y, Sato Y, Nagata M. Distribution and components of interstitial inflammation and fibrosis in IgG4-related kidney disease: analysis of autopsy specimens. Hum Pathol 2016; 55:164-73. [PMID: 27246178 DOI: 10.1016/j.humpath.2016.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/30/2016] [Accepted: 05/19/2016] [Indexed: 12/24/2022]
Abstract
IgG4-related kidney disease (IgG4-RKD) occasionally progresses to chronic renal failure and is pathologically characterized by IgG4-positive lymphoplasmacyte-rich tubulointerstitial nephritis with storiform fibrosis (bird's-eye pattern fibrosis). Although radiology reveals a heterogeneous distribution of affected areas in this disease, their true distribution within the whole kidney is still unknown because of difficulty in estimating this from needle biopsy samples. Using 5 autopsy specimens, the present study histologically characterized the distribution and components of interstitial inflammation and fibrosis in IgG4-RKD. Interstitial lymphoplasmacytic infiltration or fibrosis was observed in a variety of anatomical locations such as intracapsular, subcapsular, cortical, perivascular, and perineural regions heterogeneously in a patchy distribution. They tended to be more markedly accumulated around medium- and small-sized vessels. Storiform fibrosis was limited to the cortex. Immunostaining revealed nonfibrillar collagens (collagen IV and VI) and fibronectin predominance in the cortical lesion, including storiform fibrosis. In contrast, fibril-forming collagens (collagen I and III), collagen VI, and fibronectin were the main components in the perivascular lesion. In addition, α-smooth muscle actin-positive myofibroblasts were prominently accumulated in the early lesion and decreased with progression, suggesting that myofibroblasts produce extracellular matrices forming a peculiar fibrosis. In conclusion, perivascular inflammation or fibrosis of medium- and small-sized vessels is a newly identified pathologic feature of IgG4-RKD. Because storiform fibrosis contains mainly nonfibrillar collagens, "interstitial fibrosclerosis" would be a suitable term to reflect this. The relation between the location and components of fibrosis determined in whole kidney samples provides new clues to the pathophysiology underlying IgG4-RKD.
Collapse
Affiliation(s)
- Satoshi Hara
- Department of Kidney and Vascular Pathology, University of Tsukuba, Tsukuba 305-8575, Japan; Division of Rheumatology, Department of Internal Medicine, Kanazawa University Graduate School of Medicine, Kanazawa 920-8641, Japan
| | - Mitsuhiro Kawano
- Division of Rheumatology, Department of Internal Medicine, Kanazawa University Graduate School of Medicine, Kanazawa 920-8641, Japan.
| | - Ichiro Mizushima
- Division of Rheumatology, Department of Internal Medicine, Kanazawa University Graduate School of Medicine, Kanazawa 920-8641, Japan
| | - Kenichi Harada
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa 920-8640, Japan
| | - Takuma Takata
- Department of Internal Medicine, Nagaoka Chuo General Hospital, Nagaoka 940-8653, Japan
| | - Takako Saeki
- Department of Internal Medicine, Nagaoka Red Cross Hospital, Nagaoka 940-2108, Japan
| | - Yoshifumi Ubara
- Nephrology Center, Toranomon Hospital, Kajigaya 213-8587, Japan
| | - Yasuharu Sato
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Michio Nagata
- Department of Kidney and Vascular Pathology, University of Tsukuba, Tsukuba 305-8575, Japan
| |
Collapse
|
44
|
Wang HW, Shi L, Xu YP, Qin XY, Wang QZ. Oxymatrine inhibits renal fibrosis of obstructive nephropathy by downregulating the TGF-β1-Smad3 pathway. Ren Fail 2016; 38:945-51. [PMID: 27050799 DOI: 10.3109/0886022x.2016.1164185] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study investigated whether oxymatrine (OMT) treatment can ameliorate renal interstitial fibrosis in unilateral ureteral obstruction (UUO) mice model. Moreover, the potential mechanisms of such treatment were analyzed. Twenty-four C57/BL6 mice were randomly divided into three groups, namely sham group, vehicle plus unilateral ureteral obstruction (UUO)-treated group, and 100 mg/kg/d OMT plus UUO-treated group. All mice were euthanized seven days after surgery, and their kidneys were harvested. Renal injury, fibrosis, expression of proinflammatory cytokines, and the transforming growth factor-β1/Smads (TGF-β/Smads) and nuclear factor-kappa B (NF-κB)-signaling pathways were assessed. The results showed OMT significantly prevented kidney injury and fibrosis, as evidenced by decreased expression of collagen-1 and fibronectin. Furthermore, OMT administration inhibited the release of inflammatory factors including tumor necrosis factor-α, (TNF-α) interleukin-1β (IL-1β), and interleukin-6 (IL-6), as well as phosphorylated NF-κB p65. In addition, OMT blocked the activation of myofibroblasts by inhibiting the TGF-β/Smad3-signaling pathway. The findings indicate that OMT-attenuated renal fibrosis and inflammation, and this renoprotective effect may be ascribed to the inactivation of the TGF-β/Smad3 and NF-κB p65 pathways.
Collapse
Affiliation(s)
- Hong-Wei Wang
- a Department of Cardiology , People's Hospital of Xianfeng County , Xianfeng , China
| | - Lei Shi
- b Department of Oncology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Yan-Ping Xu
- c Department of Scientific Research Office , Renmin Hospital of Wuhan University , Wuhan , China
| | - Xing-Ya Qin
- d Department of Orthopedics , People's Hospital of Xianfeng County , Xianfeng , China
| | - Qi-Zhi Wang
- e Department of Gastroenterology , People's Hospital of Xianfeng County , Xianfeng , China
| |
Collapse
|
45
|
Zhao YL, Zhu RT, Sun YL. Epithelial-mesenchymal transition in liver fibrosis. Biomed Rep 2016; 4:269-274. [PMID: 26998262 DOI: 10.3892/br.2016.578] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/07/2016] [Indexed: 12/11/2022] Open
Abstract
Liver fibrosis is the result of a sustained wound healing response to sustained chronic liver injury, which includes viral, alcoholic and autoimmune hepatitis. Hepatic regeneration is the dominant outcome of liver damage. The outcomes of successful repair are the replacement of dead epithelial cells with healthy epithelial cells, and reconstruction of the normal hepatic structure and function. Prevention of the development of epithelial-mesenchymal transition (EMT) may control and even reverse liver fibrosis. EMT is a critical process for an epithelial cell to undergo a conversion to a mesenchymal phenotype, and is believed to be an inflammation-induced response, which may have a central role in liver fibrosis. The origin of fibrogenic cells in liver fibrosis remains controversial. Numerous studies have investigated the origin of all fibrogenic cells within the liver and the mechanism of the signaling pathways that lead to the activation of EMT programs during numerous chronic liver diseases. The present study aimed to summarize the evidence to explain the possible role of EMT in liver fibrosis.
Collapse
Affiliation(s)
- Ya-Lei Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary and Pancreatic Diseases, School of Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Rong-Tao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary and Pancreatic Diseases, School of Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Yu-Ling Sun
- Department of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary and Pancreatic Diseases, School of Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| |
Collapse
|
46
|
Ma FY, Blease K, Nikolic-Paterson DJ. A role for spleen tyrosine kinase in renal fibrosis in the mouse obstructed kidney. Life Sci 2016; 146:192-200. [PMID: 26779657 DOI: 10.1016/j.lfs.2016.01.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/28/2015] [Accepted: 01/13/2016] [Indexed: 02/08/2023]
Abstract
AIMS Spleen tyrosine kinase (Syk) is a non-receptor tyrosine kinase involved in the signalling pathways of the B cell receptor, Fcγ-receptor and some leukocyte integrins. However, Syk can also be expressed by some non-haematopoietic cell types, although whether Syk signalling in these cells contributes to the pathogenesis of kidney disease is unknown. To address this question, we examined the function of Syk in antibody-independent renal interstitial fibrosis in the unilateral ureteric obstruction (UUO) model. MAIN METHODS Groups of C57BL/6J mice were treated with a selective Syk inhibitor (CC0417, 30 mg/kg/bid), vehicle, or no treatment, from the time of surgery until being killed 7 days later. KEY FINDINGS A substantial accumulation of interstitial Syk(+) cells was seen in the UUO kidney. Double staining identified Syk expression by infiltrating macrophages and by a subset of α-SMA(+) myofibroblasts. CC0417 treatment substantially reduced the Syk(+) cell population in conjunction with a reduction in both myofibroblast and macrophage accumulation. This was associated with a substantial reduction in collagen IV deposition and mRNA levels of pro-fibrotic (collagen I, collagen IV, fibronectin, α-SMA, TGF-β1 and PAI-1) and pro-inflammatory molecules (MCP-1, TNF-α and NOS2). CC0417 treatment reduced both PDGF-B mRNA levels and Ki67(+) proliferating interstitial cells in the UUO kidney. Furthermore, CC0417 inhibited PDGF-AB induced ERK activation and cell proliferation of cultured primary kidney fibroblasts. SIGNIFICANCE This study has identified a pathologic role for Syk in renal interstitial fibrosis. Syk inhibitors may have therapeutic potential in chronic fibrotic kidney disease.
Collapse
Affiliation(s)
- Frank Y Ma
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria 3168, Australia; Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | | | - David J Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre, Monash Health, Clayton, Victoria 3168, Australia; Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria 3168, Australia.
| |
Collapse
|
47
|
Mao S, Zhang J. The emerging role of hepatocyte growth factor in renal diseases. J Recept Signal Transduct Res 2015; 36:303-9. [DOI: 10.3109/10799893.2015.1080275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Loeffler I, Wolf G. Epithelial-to-Mesenchymal Transition in Diabetic Nephropathy: Fact or Fiction? Cells 2015; 4:631-52. [PMID: 26473930 PMCID: PMC4695850 DOI: 10.3390/cells4040631] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 12/17/2022] Open
Abstract
The pathophysiology of diabetic nephropathy (DN), one of the most serious complications in diabetic patients and the leading cause of end-stage renal disease worldwide, is complex and not fully elucidated. A typical hallmark of DN is the excessive deposition of extracellular matrix (ECM) proteins in the glomerulus and in the renal tubulointerstitium, eventually leading to glomerulosclerosis and interstitial fibrosis. Although it is obvious that myofibroblasts play a major role in the synthesis and secretion of ECM, the origin of myofibroblasts in DN remains the subject of controversial debates. A number of studies have focused on epithelial-to-mesenchymal transition (EMT) as one source of matrix-generating fibroblasts in the diseased kidney. EMT is characterized by the acquisition of mesenchymal properties by epithelial cells, preferentially proximal tubular cells and podocytes. In this review we comprehensively review the literature and discuss arguments both for and against a function of EMT in renal fibrosis in DN. While the precise extent of the contribution to nephrotic fibrosis is certainly arduous to quantify, the picture that emerges from this extensive body of literature suggests EMT as a major source of myofibroblasts in DN.
Collapse
Affiliation(s)
- Ivonne Loeffler
- Department of Internal Medicine III, University Hospital, University of Jena, Erlanger Allee 101, D-07747 Jena, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital, University of Jena, Erlanger Allee 101, D-07747 Jena, Germany.
| |
Collapse
|
49
|
Liu T, Nie F, Yang X, Wang X, Yuan Y, Lv Z, Zhou L, Peng R, Ni D, Gu Y, Zhou Q, Weng Y. MicroRNA-590 is an EMT-suppressive microRNA involved in the TGFβ signaling pathway. Mol Med Rep 2015; 12:7403-11. [PMID: 26459119 PMCID: PMC4626157 DOI: 10.3892/mmr.2015.4374] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 09/09/2015] [Indexed: 01/01/2023] Open
Abstract
Over the last few decades, the epithelial-to-mesenchymal transition (EMT) has been identified as being involved in a number of aspects of physiological processes and various pathological events, including embryonic development and renal fibrosis. Transforming growth factor-β receptor 2 (TGFβR2) is a widely studied gene, which fulfils a vital role in the TGFβ signaling pathway and exerts a crucial function in the progression of EMT. Previous studies demonstrated that the dysregulation of microRNAs (miRNAs) is considered to be associated with the EMT process. However, the precise functional involvement of miRNAs in EMT remains to be fully elucidated. In the present study, the level of miR-590 was decreased in an EMT model in vitro and in vivo. Furthermore, the overexpression of miR-590 inhibited EMT by upregulating the epithelial marker, E-cadherin, and downregulating the mesenchymal markers, laminin, α-smooth muscle actin (α-SMA) and collagen, in the human kidney 2 (HK2) cell line. Furthermore, TGFβR2 was negatively regulated by miR-590. In addition, performing a knockdown of TGFβR2 with small-interfering RNA had an effect similar to miR-590 on EMT in the HK2 cell line, whereas the transfection of pCMV-tag2B-TGFβR2 reversed the effect of miR-590 on EMT in HK2 cells. Taken together, the present study demonstrated that miR-590 is a novel EMT-suppressive microRNA, which targets TGFβR2.
Collapse
Affiliation(s)
- Tianming Liu
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Fang Nie
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xianggui Yang
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaoyan Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yue Yuan
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhongshi Lv
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Li Zhou
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Rui Peng
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Dongsheng Ni
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yuping Gu
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qin Zhou
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yaguang Weng
- Key Laboratory of Laboratory Medical Diagnostics Designated by Chinese Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
50
|
Li M, Luan F, Zhao Y, Hao H, Zhou Y, Han W, Fu X. Epithelial-mesenchymal transition: An emerging target in tissue fibrosis. Exp Biol Med (Maywood) 2015; 241:1-13. [PMID: 26361988 DOI: 10.1177/1535370215597194] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 06/19/2015] [Indexed: 12/18/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is involved in a variety of tissue fibroses. Fibroblasts/myofibroblasts derived from epithelial cells contribute to the excessive accumulation of fibrous connective tissue in damaged tissue, which can lead to permanent scarring or organ malfunction. Therefore, EMT-related fibrosis cannot be neglected. This review highlights the findings that demonstrate the EMT to be a direct contributor to the fibroblast/myofibroblast population in the development of tissue fibrosis and helps to elucidate EMT-related anti-fibrotic strategies, which may enable the development of therapeutic interventions to suppress EMT and potentially reverse organ fibrosis.
Collapse
Affiliation(s)
- Meirong Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya 572014, P. R. China
| | - Fuxin Luan
- Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya 572014, P. R. China
| | - Yali Zhao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya 572014, P. R. China
| | - Haojie Hao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Yong Zhou
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Weidong Han
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing 100853, P. R. China
| |
Collapse
|