201
|
Hao H, Li X, Li Q, Lin H, Chen Z, Xie J, Xuan W, Liao W, Bin J, Huang X, Kitakaze M, Liao Y. FGF23 promotes myocardial fibrosis in mice through activation of β-catenin. Oncotarget 2018; 7:64649-64664. [PMID: 27579618 PMCID: PMC5323105 DOI: 10.18632/oncotarget.11623] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/21/2016] [Indexed: 11/25/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) has been reported to induce left ventricular hypertrophy, but it remains unclear whether FGF23 plays a role in cardiac fibrosis. This study is attempted to investigate the role of FGF23 in post-infarct myocardial fibrosis in mice. We noted that myocardial and plasma FGF23 and FGF receptor 4 were increased in mice with heart failure as well as in cultured adult mouse cardiac fibroblasts (AMCFs) exposed to angiotensin II, phenylephrine, soluble fractalkine. Recombinant FGF23 protein increased active β-catenin , procollagen I and procollagen III expression in cultured AMCFs. Furthermore, intra-myocardial injection of adeno-associated virus-FGF23 in mice significantly increased left ventricular end-diastolic pressure and myocardial fibrosis, and markedly upregulated active β-catenin, transforming growth factor β (TGF-β), procollagen I and procollagen III in both myocardial infarction (MI) and ischemia/reperfusion (IR) mice, while β-catenin inhibitor or silencing of β-catenin antagonized the FGF23-promoted myocardial fibrosis in vitro and in vivo. These findings indicate that FGF23 promotes myocardial fibrosis and exacerbates diastolic dysfunction induced by MI or IR, which is associated with the upregulation of active β-catenin and TGF-β.
Collapse
Affiliation(s)
- Huixin Hao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xixian Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingman Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hairuo Lin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenhuan Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiahe Xie
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wanling Xuan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaobo Huang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Masafumi Kitakaze
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Cardiovascular Division of the Department of Medicine, National Cerebral and Cardiovascular Center, Fujishirodai, Suita, Osaka, Japan
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
202
|
Luo C, Zhou S, Zhou Z, Liu Y, Yang L, Liu J, Zhang Y, Li H, Liu Y, Hou FF, Zhou L. Wnt9a Promotes Renal Fibrosis by Accelerating Cellular Senescence in Tubular Epithelial Cells. J Am Soc Nephrol 2018; 29:1238-1256. [PMID: 29440280 DOI: 10.1681/asn.2017050574] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 01/03/2018] [Indexed: 12/24/2022] Open
Abstract
Cellular senescence is associated with renal disease progression, and accelerated tubular cell senescence promotes the pathogenesis of renal fibrosis. However, the underlying mechanism is unknown. We assessed the potential role of Wnt9a in tubular cell senescence and renal fibrosis. Compared with tubular cells of normal subjects, tubular cells of humans with a variety of nephropathies and those of several mouse models of CKD expressed high levels of Wnt9a that colocalized with the senescence-related protein p16INK4A Wnt9a expression level correlated with the extent of renal fibrosis, decline of eGFR, and expression of p16INK4A Furthermore, ectopic expression of Wnt9a after ischemia-reperfusion injury (IRI) induced activation of β-catenin and exacerbated renal fibrosis. Overexpression of Wnt9a exacerbated tubular senescence, evidenced by increased detection of p16INK4A expression and senescence-associated β-galactosidase activity. Conversely, shRNA-mediated knockdown of Wnt9a repressed IRI-induced renal fibrosis in vivo and impeded the growth of senescent tubular epithelial cells in culture. Notably, Wnt9a-induced renal fibrosis was inhibited by shRNA-mediated silencing of p16INK4A in the IRI mouse model. In a human proximal tubular epithelial cell line and primary renal tubular cells, Wnt9a remarkably upregulated levels of senescence-related p16INK4A, p19ARF, p53, and p21 and decreased the phosphorylation of retinoblastoma protein. Wnt9a also induced senescent tubular cells to produce TGF-β1, which promoted proliferation and activation in normal rat kidney fibroblasts. Thus, Wnt9a drives tubular senescence and fibroblast activation. Furthermore, the Wnt9a-TGF-β pathway appears to create a reciprocal activation loop between senescent tubular cells and activated fibroblasts that promotes and accelerates the pathogenesis of renal fibrosis.
Collapse
Affiliation(s)
- Congwei Luo
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and
| | - Zhanmei Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and
| | - Yahong Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and
| | - Li Yang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and
| | - Jiafeng Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and
| | - Yunfang Zhang
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China; and
| | - Hongyan Li
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China; and
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital and
| |
Collapse
|
203
|
The role of fibroblast growth factor 23 and Klotho in uremic cardiomyopathy. Curr Opin Nephrol Hypertens 2018; 25:314-24. [PMID: 27219043 DOI: 10.1097/mnh.0000000000000231] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW In chronic kidney disease (CKD), multiple factors contribute to the development of cardiac hypertrophy by directly targeting the heart or indirectly by inducing systemic changes such as hypertension, anemia, and inflammation. Furthermore, disturbances in phosphate metabolism have been identified as nonclassical risk factors for cardiovascular mortality in these patients. With declining kidney function, the physiologic regulators of phosphate homeostasis undergo changes in their activity as well as their circulating levels, thus potentially contributing to cardiac hypertrophy once they are out of balance. Recently, two of these phosphate regulators, fibroblast growth factor 23 (FGF23) and Klotho, have been shown to affect cardiac remodeling, thereby unveiling a novel pathomechanism of cardiac hypertrophy in CKD. Here we discuss the potential direct versus indirect effects of FGF23 and the soluble form of Klotho on the heart, and their crosstalk in the regulation of cardiac hypertrophy. RECENT FINDINGS In models of CKD, FGF23 can directly target cardiac myocytes via FGF receptor 4 and induce cardiac hypertrophy in a blood pressure-independent manner. Soluble Klotho may directly target the heart via an unknown receptor thereby protecting the myocardium from pathologic stress stimuli that are associated with CKD, such as uremic toxins or FGF23. SUMMARY Elevated serum levels of FGF23 and reduced serum levels of soluble Klotho contribute to uremic cardiomyopathy in a synergistic manner.
Collapse
|
204
|
Zhou D, Fu H, Xiao L, Mo H, Zhuo H, Tian X, Lin L, Xing J, Liu Y. Fibroblast-Specific β-Catenin Signaling Dictates the Outcome of AKI. J Am Soc Nephrol 2018; 29:1257-1271. [PMID: 29343518 DOI: 10.1681/asn.2017080903] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 12/18/2017] [Indexed: 01/10/2023] Open
Abstract
AKI is a devastating condition with high morbidity and mortality. The pathologic features of AKI are characterized by tubular injury, inflammation, and vascular impairment. Whether fibroblasts in the renal interstitium have a role in the pathogenesis of AKI is unknown. In this study, we investigated the role of fibroblast-specific β-catenin signaling in dictating the outcome of AKI, using conditional knockout mice in which β-catenin was specifically ablated in fibroblasts (Gli1-β-cat-/-). After ischemia-reperfusion injury (IRI), Gli1-β-cat-/- mice had lower serum creatinine levels and less morphologic injury than Gli1-β-cat+/+ littermate controls. Moreover, we detected fewer apoptotic cells, as well as decreased cytochrome C release; reduced expression of Bax, FasL, and p53; and increased phosphorylation of Akt, in the Gli1-β-cat-/- kidneys. Gli1-β-cat-/- kidneys also exhibited upregulated expression of proliferating cell nuclear antigen and Ki-67, which are markers of cell proliferation. Furthermore, Gli1-β-cat-/- kidneys displayed suppressed NF-κB signaling and cytokine expression and reduced infiltration of inflammatory cells. Notably, loss of β-catenin in fibroblasts induced renal expression of hepatocyte growth factor (HGF) and augmented the tyrosine phosphorylation of c-met receptor after IRI. In vitro, treatment with Wnt ligands or ectopic expression of active β-catenin inhibited HGF mRNA and protein expression and repressed HGF promoter activity. Collectively, these results suggest that fibroblast-specific β-catenin signaling can control tubular injury and repair in AKI by modulating HGF expression. Our studies uncover a previously unrecognized role for interstitial fibroblasts in the pathogenesis of AKI.
Collapse
Affiliation(s)
| | - Haiyan Fu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liangxiang Xiao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | - Xiaojun Tian
- Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Lin Lin
- Departments of Pathology and
| | - Jianhua Xing
- Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Youhua Liu
- Departments of Pathology and .,State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
205
|
Abstract
Aging impacts diseases and lifespan. With current knowledge of stem cells, it is feasible to design and test interventions that delay aging and improve both health and lifespan. Stem cells, together with anti-aging genes such as Klotho, play a crucial role in delaying the aging process. Stem cells in combination with anti-aging genes make a complex and protective shield, which stand against the eroding effects of aging. Increased wear and tear of the stem cells, as well as Klotho deficiency, is expected to heavily increase cellular damage and accelerate the process of aging. Stem cells in conjugation with anti-aging genes probably receive and neutralize most of the devastating signaling effects which are known to cause premature aging. The shield of stem cells combined with anti-aging genes is a primary target for absorbing the shock of aging. If this shield neutralizes the shocks, it could lead to a youthful state, but if not it will accelerate the aging journey. In this review, we concisely discuss the neutralizing ability, operated and regulated by stem cells and other life-extension factors. We suggest that stem cell interventions that increase rejuvenation and keep in balance the expression of anti-aging genes could delay the aging phenotypes and result in prolonged lifespan.
Collapse
Affiliation(s)
- Mujib Ullah
- Department of Physiology, BMSB, College of Medicine, University of Oklahoma Health Sciences Center (OUHSC), 940 Stanton L. Young Blvd, Oklahoma City, OK, 73126-0901, USA.
| | - Zhongjie Sun
- Department of Physiology, BMSB, College of Medicine, University of Oklahoma Health Sciences Center (OUHSC), 940 Stanton L. Young Blvd, Oklahoma City, OK, 73126-0901, USA
| |
Collapse
|
206
|
Takenaka T, Inoue T, Miyazaki T, Kobori H, Nishiyama A, Ishii N, Hayashi M, Suzuki H. Klotho suppresses the renin-angiotensin system in adriamycin nephropathy. Nephrol Dial Transplant 2018; 32:791-800. [PMID: 27798196 DOI: 10.1093/ndt/gfw340] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/10/2016] [Indexed: 11/13/2022] Open
Abstract
Backgrounds Klotho protein interacts with the transforming growth factor β (TGF-β) receptor and Wnt, which contribute to the progression of renal disease, inhibiting their signals. Renal and circulating klotho levels are diminished in chronic kidney disease. Methods Experiments were performed to assess whether supplementation of klotho protein could have protective effects on the kidney. Rats were injected with adriamycin (5 mg/kg) and divided into three groups: those treated with vehicle, those treated with klotho protein and those treated with klotho plus 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD). Rats without adriamycin treatment were used as a control. Results Adriamycin reduced the serum klotho concentration and renal expression of klotho and E-cadherin. Adriamycin also increased the renal expression of Wnt, TGF-β, and angiotensinogen, as well as the renal abundance of β-catenin and angiotensin II. Klotho supplementation suppressed adriamycin-induced elevations of β-catenin and angiotensin II with sustained Wnt expression. Combined treatment with klotho and TDZD reversed the klotho-induced improvements in the renal abundance of β-catenin and angiotensin II as well as the expression of TGF-β and angiotensinogen without affecting E-cadherin. Conclusions Our data indicate that Wnt is involved in the pathogenesis of adriamycin nephropathy. Furthermore, klotho supplementation inhibited Wnt signaling, ameliorating renal angiotensin II. Finally, klotho protein appears to suppress epithelial-mesenchymal transition by inhibiting TGF-β and Wnt signaling.
Collapse
Affiliation(s)
- Tsuneo Takenaka
- Department of Medicine, International University of Health and Welfare, 8-10-16 Akasaka, Minato, Tokyo 107-0052, Japan
| | - Tsutomu Inoue
- Department of Nephrology, Saitama Medical University, Iruma, Saitama, Japan
| | - Takashi Miyazaki
- Department of Nephrology, Saitama Medical University, Iruma, Saitama, Japan
| | - Hiroyuki Kobori
- Department of Medicine, International University of Health and Welfare, 8-10-16 Akasaka, Minato, Tokyo 107-0052, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kida, Kagawa, Japan
| | - Naohito Ishii
- Department of Clinical Chemistry, Kitasato University, Sagamihara, Kanagawa, Japan
| | | | - Hiromichi Suzuki
- Department of Nephrology, Saitama Medical University, Iruma, Saitama, Japan
| |
Collapse
|
207
|
Li SS, He AL, Deng ZY, Liu QF. Ginsenoside-Rg1 Protects against Renal Fibrosis by Regulating the Klotho/TGF-β1/Smad Signaling Pathway in Rats with Obstructive Nephropathy. Biol Pharm Bull 2018; 41:585-591. [DOI: 10.1248/bpb.b17-00934] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Sha-sha Li
- Clinical Research & Lab Centre, Kunshan First People’s Hospital Affiliated to Jiangsu University
| | - Ao-lin He
- Clinical Research & Lab Centre, Kunshan First People’s Hospital Affiliated to Jiangsu University
| | - Zhi-yong Deng
- Department of Pathology, Kunshan First People’s Hospital Affiliated to Jiangsu University
| | - Qi-feng Liu
- Department of Nephrology, Kunshan First People’s Hospital Affiliated to Jiangsu University
| |
Collapse
|
208
|
Wang Y, Zhou CJ, Liu Y. Wnt Signaling in Kidney Development and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 153:181-207. [PMID: 29389516 PMCID: PMC6008255 DOI: 10.1016/bs.pmbts.2017.11.019] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Wnt signal cascade is an evolutionarily conserved, developmental pathway that regulates embryogenesis, injury repair, and pathogenesis of human diseases. It is well established that Wnt ligands transmit their signal via canonical, β-catenin-dependent and noncanonical, β-catenin-independent mechanisms. Mounting evidence has revealed that Wnt signaling plays a key role in controlling early nephrogenesis and is implicated in the development of various kidney disorders. Dysregulations of Wnt expression cause a variety of developmental abnormalities and human diseases, such as congenital anomalies of the kidney and urinary tract, cystic kidney, and renal carcinoma. Multiple Wnt ligands, their receptors, and transcriptional targets are upregulated during nephron formation, which is crucial for mediating the reciprocal interaction between primordial tissues of ureteric bud and metanephric mesenchyme. Renal cysts are also associated with disrupted Wnt signaling. In addition, Wnt components are important players in renal tumorigenesis. Activation of Wnt/β-catenin is instrumental for tubular repair and regeneration after acute kidney injury. However, sustained activation of this signal cascade is linked to chronic kidney diseases and renal fibrosis in patients and experimental animal models. Mechanistically, Wnt signaling controls a diverse array of biologic processes, such as cell cycle progression, cell polarity and migration, cilia biology, and activation of renin-angiotensin system. In this chapter, we have reviewed recent findings that implicate Wnt signaling in kidney development and diseases. Targeting this signaling may hold promise for future treatment of kidney disorders in patients.
Collapse
Affiliation(s)
- Yongping Wang
- National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Chengji J Zhou
- University of California Davis, Sacramento, CA, United States
| | - Youhua Liu
- National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| |
Collapse
|
209
|
Richter B, Faul C. FGF23 Actions on Target Tissues-With and Without Klotho. Front Endocrinol (Lausanne) 2018; 9:189. [PMID: 29770125 PMCID: PMC5940753 DOI: 10.3389/fendo.2018.00189] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 is a phosphaturic hormone whose physiologic actions on target tissues are mediated by FGF receptors (FGFR) and klotho, which functions as a co-receptor that increases the binding affinity of FGF23 for FGFRs. By stimulating FGFR/klotho complexes in the kidney and parathyroid gland, FGF23 reduces renal phosphate uptake and secretion of parathyroid hormone, respectively, thereby acting as a key regulator of phosphate metabolism. Recently, it has been shown that FGF23 can also target cell types that lack klotho. This unconventional signaling event occurs in an FGFR-dependent manner, but involves other downstream signaling pathways than in "classic" klotho-expressing target organs. It appears that klotho-independent signaling mechanisms are only activated in the presence of high FGF23 concentrations and result in pathologic cellular changes. Therefore, it has been postulated that massive elevations in circulating levels of FGF23, as found in patients with chronic kidney disease, contribute to associated pathologies by targeting cells and tissues that lack klotho. This includes the induction of cardiac hypertrophy and fibrosis, the elevation of inflammatory cytokine expression in the liver, and the inhibition of neutrophil recruitment. Here, we describe the signaling and cellular events that are caused by FGF23 in tissues lacking klotho, and we discuss FGF23's potential role as a hormone with widespread pathologic actions. Since the soluble form of klotho can function as a circulating co-receptor for FGF23, we also discuss the potential inhibitory effects of soluble klotho on FGF23-mediated signaling which might-at least partially-underlie the pleiotropic tissue-protective functions of klotho.
Collapse
|
210
|
Hu F, Xue M, Li Y, Jia YJ, Zheng ZJ, Yang YL, Guan MP, Sun L, Xue YM. Early Growth Response 1 (Egr1) Is a Transcriptional Activator of NOX4 in Oxidative Stress of Diabetic Kidney Disease. J Diabetes Res 2018; 2018:3405695. [PMID: 29854821 PMCID: PMC5944279 DOI: 10.1155/2018/3405695] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/11/2017] [Accepted: 11/21/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND NADPH oxidase 4 (NOX4) plays a major role in renal oxidative stress of diabetic kidney disease (DKD). NOX4 was significantly increased in Egr1-expressing fibroblasts, but the relationship between Egr1 and NOX4 in DKD is unclear. METHODS For the evaluation of the potential relationship between Egr1 and NOX4, both were detected in HFD/STZ-induced mice and HK-2 cells treated with TGF-β1. Then, changes in NOX4 expression were detected in HK-2 cells and mice with overexpression and knockdown of Egr1. The direct relationship between Egr1 and NOX4 was explored via chromatin immunoprecipitation (ChIP). RESULTS We found increased levels of Egr1, NOX4, and α-SMA in the kidney cortices of diabetic mice and in TGF-β1-treated HK-2 cells. Overexpression or silencing of Egr1 in HK-2 cells could upregulate or downregulate NOX4 and α-SMA. ChIP assays revealed that TGF-β1 induced Egr1 to bind to the NOX4 promoter. Finally, Egr1 overexpression or knockdown in diabetic mice could upregulate or downregulate the expression of NOX4 and ROS, and α-SMA was also changed. CONCLUSION Our study provides strong evidence that Egr1 is a transcriptional activator of NOX4 in oxidative stress of DKD. Egr1 contributes to DKD by enhancing EMT, in part by targeting NOX4.
Collapse
Affiliation(s)
- Fang Hu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Meng Xue
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Endocrinology and Metabolism, Shenzhen People's Hospital, Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Yang Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Geriatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi-Jie Jia
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zong-Ji Zheng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Lin Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mei-Ping Guan
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Liao Sun
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Yao-Ming Xue
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
211
|
Leifheit-Nestler M, Haffner D. Paracrine Effects of FGF23 on the Heart. Front Endocrinol (Lausanne) 2018; 9:278. [PMID: 29892269 PMCID: PMC5985311 DOI: 10.3389/fendo.2018.00278] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/11/2018] [Indexed: 12/17/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 is a phosphaturic hormone primarily secreted by osteocytes to maintain phosphate and mineral homeostasis. In patients with and without chronic kidney disease, enhanced circulating FGF23 levels associate with pathologic cardiac remodeling, i.e., left ventricular hypertrophy (LVH) and myocardial fibrosis and increased cardiovascular mortality. Experimental studies demonstrate that FGF23 promotes hypertrophic growth of cardiac myocytes via FGF receptor 4-dependent activation of phospholipase Cγ/calcineurin/nuclear factor of activated T cell signaling independent of its co-receptor klotho. Recent studies indicate that FGF23 is also expressed in the heart, and markedly enhanced in various clinical and experimental settings of cardiac remodeling and heart failure independent of preserved or reduced renal function. On a cellular level, FGF23 is expressed in cardiac myocytes and in other non-cardiac myocytes, including cardiac fibroblasts, vascular smooth muscle and endothelial cells in coronary arteries, and in inflammatory macrophages. Current data suggest that secreted by cardiac myocytes, FGF23 can stimulate pro-fibrotic factors in myocytes to induce fibrosis-related pathways in fibroblasts and consequently cardiac fibrosis in a paracrine manner. While acting on cardiac myocytes, FGF23 directly induces pro-hypertrophic genes and promotes the progression of LVH in an autocrine and paracrine fashion. Thus, enhanced FGF23 may promote cardiac injury in various clinical settings not only by endocrine but also via paracrine/autocrine mechanisms. In this review, we discuss recent clinical and experimental data regarding molecular mechanisms of FGF23's paracrine action on the heart with respect to pathological cardiac remodeling.
Collapse
|
212
|
Thiagarajan D, O’ Shea K, Sreejit G, Ananthakrishnan R, Quadri N, Li Q, Schmidt AM, Gabbay K, Ramasamy R. Aldose reductase modulates acute activation of mesenchymal markers via the β-catenin pathway during cardiac ischemia-reperfusion. PLoS One 2017; 12:e0188981. [PMID: 29190815 PMCID: PMC5708684 DOI: 10.1371/journal.pone.0188981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022] Open
Abstract
Aldose reductase (AR: human, AKR1B1; mouse, AKR1B3), the first enzyme in the polyol pathway, plays a key role in mediating myocardial ischemia/reperfusion (I/R) injury. In earlier studies, using transgenic mice broadly expressing human AKR1B1 to human-relevant levels, mice devoid of Akr1b3, and pharmacological inhibitors of AR, we demonstrated that AR is an important component of myocardial I/R injury and that inhibition of this enzyme protects the heart from I/R injury. In this study, our objective was to investigate if AR modulates the β-catenin pathway and consequent activation of mesenchymal markers during I/R in the heart. To test this premise, we used two different experimental models: in vivo, Akr1b3 null mice and wild type C57BL/6 mice (WT) were exposed to acute occlusion of the left anterior descending coronary artery (LAD) followed by recovery for 48 hours or 28 days, and ex-vivo, WT and Akr1b3 null murine hearts were perfused using the Langendorff technique (LT) and subjected to 30 min of global (zero-flow) ischemia followed by 60 min of reperfusion. Our in vivo results reveal reduced infarct size and improved functional recovery at 48 hours in mice devoid of Akr1b3 compared to WT mice. We demonstrate that the cardioprotection observed in Akr1b3 null mice was linked to acute activation of the β-catenin pathway and consequent activation of mesenchymal markers and genes linked to fibrotic remodeling. The increased activity of the β-catenin pathway at 48 hours of recovery post-LAD was not observed at 28 days post-infarction, thus indicating that the observed increase in β-catenin activity was transient in the mice hearts devoid of Akr1b3. In ex vivo studies, inhibition of β-catenin blocked the cardioprotection observed in Akr1b3 null mice hearts. Taken together, these data indicate that AR suppresses acute activation of β-catenin and, thereby, blocks consequent induction of mesenchymal markers during early reperfusion after myocardial ischemia. Inhibition of AR might provide a therapeutic opportunity to optimize cardiac remodeling after I/R injury.
Collapse
Affiliation(s)
- Devi Thiagarajan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Karen O’ Shea
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Gopalkrishna Sreejit
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Radha Ananthakrishnan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Nosirudeen Quadri
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Qing Li
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Kenneth Gabbay
- Department of Pediatrics, Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
213
|
Jaturakan O, Buranakarl C, Dissayabutra T, Chaiyabutr N, Kijtawornrat A, Rungsipipat A. Changes of Klotho protein and Klotho mRNA expression in a hydroxy-L-proline induced hyperoxaluric rat model. J Vet Med Sci 2017; 79:1861-1869. [PMID: 28943597 PMCID: PMC5709566 DOI: 10.1292/jvms.17-0340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Klotho protein is recognized as having a renoprotective effect and is used as a biomarker for kidney injury. We investigated the level of Klotho protein in hyperoxaluria-induced kidney injury and the effects of vitamin E (Vit E)
and vitamin C (Vit C) supplementation. Hyperoxaluria was induced by feeding 2% (w/v) Hydroxy-L-proline (HLP) in the drinking water for 21 days. Rats were divided into 5 groups; control (Group 1, n=7), HLP treated rats that
received nothing else (Group 2, n=7), Vit E (Group 3, n=6), Vit C (Group 4, n=6) and both Vit E and Vit C (Group 5, n=7). Vit E (200 mg/kg) was injected on days 1, 6, 11 and 16, while Vit C (500 mg/kg) was given intravenously on
days 1 and 11. The Klotho protein levels and oxidative status were measured. The expression level of kidney Klotho protein expression was significantly reduced by HLP-treatment, while the mRNA expression was higher
(P<0.05), the plasma and kidney malondialdehyde and kidney superoxide dismutase activities were increased, and the kidney reduced glutathione and urinary total antioxidant status were decreased
(P<0.05). All of these changes were ameliorated by administration of Vit E, Vit C or especially the co-administration of both. In conclusion, HLP-induced hyperoxaluria reduced the kidney Klotho protein level,
which could be restored by Vit E and/or Vit C.
Collapse
Affiliation(s)
- Orapun Jaturakan
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| | - Chollada Buranakarl
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| | - Thasinas Dissayabutra
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Narongsak Chaiyabutr
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| | - Anusak Kijtawornrat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| | - Anudep Rungsipipat
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
214
|
Mencke R, Olauson H, Hillebrands JL. Effects of Klotho on fibrosis and cancer: A renal focus on mechanisms and therapeutic strategies. Adv Drug Deliv Rev 2017; 121:85-100. [PMID: 28709936 DOI: 10.1016/j.addr.2017.07.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/28/2017] [Accepted: 07/07/2017] [Indexed: 12/21/2022]
Abstract
Klotho is a membrane-bound protein predominantly expressed in the kidney, where it acts as a permissive co-receptor for Fibroblast Growth Factor 23. In its shed form, Klotho exerts anti-fibrotic effects in several tissues. Klotho-deficient mice spontaneously develop fibrosis and Klotho deficiency exacerbates the disease progression in fibrotic animal models. Furthermore, Klotho overexpression or supplementation protects against fibrosis in various models of renal and cardiac fibrotic disease. These effects are mediated at least partially by the direct inhibitory effects of soluble Klotho on TGFβ1 signaling, Wnt signaling, and FGF2 signaling. Soluble Klotho, as present in the circulation, appears to be the primary mediator of anti-fibrotic effects. Similarly, through inhibition of the TGFβ1, Wnt, FGF2, and IGF1 signaling pathways, Klotho also inhibits tumorigenesis. The Klotho promoter gene is generally hypermethylated in cancer, and overexpression or supplementation of Klotho has been found to inhibit tumor growth in various animal models. This review focuses on the protective effects of soluble Klotho in inhibiting renal fibrosis and fibrosis in distant organs secondary to renal Klotho deficiency. We also discuss the structure-function relationships of Klotho domains and biological effects in the context of potential targeted treatment strategies.
Collapse
Affiliation(s)
- Rik Mencke
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hannes Olauson
- Department of Clinical Science, Intervention and Technology (Division of Renal Medicine), Karolinska Institutet, Stockholm, Sweden
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
215
|
Laszczyk AM, Fox-Quick S, Vo HT, Nettles D, Pugh PC, Overstreet-Wadiche L, King GD. Klotho regulates postnatal neurogenesis and protects against age-related spatial memory loss. Neurobiol Aging 2017; 59:41-54. [PMID: 28837861 PMCID: PMC5612914 DOI: 10.1016/j.neurobiolaging.2017.07.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/22/2017] [Accepted: 07/21/2017] [Indexed: 12/29/2022]
Abstract
Although the absence of the age-regulating klotho protein causes klotho-deficient mice to rapidly develop cognitive impairment and increasing klotho enhances hippocampal-dependent memory, the cellular effects of klotho that mediate hippocampal-dependent memory function are unknown. Here, we show premature aging of the klotho-deficient hippocampal neurogenic niche as evidenced by reduced numbers of neural stem cells, decreased proliferation, and impaired maturation of immature neurons. Klotho-deficient neurospheres show reduced proliferation and size that is rescued by supplementation with shed klotho protein. Conversely, 6-month-old klotho-overexpressing mice exhibit increased numbers of neural stem cells, increased proliferation, and more immature neurons with enhanced dendritic arborization. Protection from normal age-related loss of object location memory with klotho overexpression and loss of spatial memory when klotho is reduced by even half suggests direct, local effects of the protein. Together, these data show that klotho is a novel regulator of postnatal neurogenesis affecting neural stem cell proliferation and maturation sufficient to impact hippocampal-dependent spatial memory function.
Collapse
Affiliation(s)
- Ann M Laszczyk
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephanie Fox-Quick
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dailey Nettles
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Phyllis C Pugh
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
216
|
Liu QF, Ye JM, Yu LX, He AL, Sun Q, He DW, Li SS. Plasma s-Klotho is related to kidney function and predicts adverse renal outcomes in patients with advanced chronic kidney disease. J Investig Med 2017; 66:669-675. [PMID: 29061648 DOI: 10.1136/jim-2017-000560] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2017] [Indexed: 12/13/2022]
Abstract
To investigate whether the soluble Klotho (s-Klotho) level in patients with chronic kidney disease (CKD) is related to kidney function and whether a low s-Klotho level can predict adverse renal outcomes or CKD progression in patients with advanced CKD. 112 patients with CKD stages 3-5 and 30 healthy volunteers were enrolled. Blood samples were collected to measure serum creatinine, calcium, phosphorus, intact parathyroid hormone, and hemoglobin. s-Klotho and fibroblast growth factor 23 (FGF23) were determined by ELISA. We first conducted a cross-sectional study to investigate correlations between s-Klotho and estimated glomerular filtration rate (eGFR) and other parameters. Patients were then followed prospectively for 20.1±10.1 months according to s-Klotho median level until serum creatinine doubled, or initiation of renal replacement therapy, or death. s-Klotho levels inpatients with CKD were significantly lower than that in the control group. For patients with CKD, there were no differences in age distribution among subgroups. However, s-Klotho level differed significantly across CKD stages, and it was lower in the advanced CKD group compared with the moderate CKD group. Correlation analysis revealed that s-Klotho was positively associated with eGFR, but inversely associated with FGF23. During the follow-up of 20.1±10.1 months, patients with higher s-Klotho levels showed a reduced risk of kidney adverse outcomes, including serum creatinine doubling and initiation of renal replacement therapy. Cox regression analysis revealed that low s-Klotho was an independent risk factor for CKD progression. s-Klotho level was closely correlated with kidney function, further, low s-Klotho level could predict adverse kidney disease outcomes in patients with progressive CKD.
Collapse
Affiliation(s)
- Qi-Feng Liu
- Department of Nephrology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Jian-Ming Ye
- Department of Nephrology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Li-Xia Yu
- Department of Nephrology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Ao-Lin He
- Clinical and Lab Research Centre, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Qiang Sun
- Department of Nephrology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Da-Wei He
- Clinical and Lab Research Centre, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| | - Sha-Sha Li
- Clinical and Lab Research Centre, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu, China
| |
Collapse
|
217
|
Mencke R, Harms G, Moser J, van Meurs M, Diepstra A, Leuvenink HG, Hillebrands JL. Human alternative Klotho mRNA is a nonsense-mediated mRNA decay target inefficiently spliced in renal disease. JCI Insight 2017; 2:94375. [PMID: 29046474 DOI: 10.1172/jci.insight.94375] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/14/2017] [Indexed: 12/11/2022] Open
Abstract
Klotho is a renal protein involved in phosphate homeostasis, which is downregulated in renal disease. It has long been considered an antiaging factor. Two Klotho gene transcripts are thought to encode membrane-bound and secreted Klotho. Indeed, soluble Klotho is detectable in bodily fluids, but the relative contributions of Klotho secretion and of membrane-bound Klotho shedding are unknown. Recent advances in RNA surveillance reveal that premature termination codons, as present in alternative Klotho mRNA (for secreted Klotho), prime mRNAs for degradation by nonsense-mediated mRNA decay (NMD). Disruption of NMD led to accumulation of alternative Klotho mRNA, indicative of normally continuous degradation. RNA IP for NMD core factor UPF1 resulted in enrichment for alternative Klotho mRNA, which was also not associated with polysomes, indicating no active protein translation. Alternative Klotho mRNA transcripts colocalized with some P bodies, where NMD transcripts are degraded. Moreover, we could not detect secreted Klotho in vitro. These results suggest that soluble Klotho is likely cleaved membrane-bound Klotho only. Furthermore, we found that, especially in acute kidney injury, splicing of the 2 mRNA transcripts is dysregulated, which was recapitulated by various noxious stimuli in vitro. This likely constitutes a novel mechanism resulting in the downregulation of membrane-bound Klotho.
Collapse
Affiliation(s)
- Rik Mencke
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.,The NIGRAM consortium detailed in the Supplemental Acknowledgments
| | - Geert Harms
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.,The NIGRAM consortium detailed in the Supplemental Acknowledgments
| | - Jill Moser
- Department of Intensive Care Medicine.,Department of Pathology and Medical Biology (Division of Medical Biology), and
| | - Matijs van Meurs
- Department of Intensive Care Medicine.,Department of Pathology and Medical Biology (Division of Medical Biology), and
| | - Arjan Diepstra
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Henri G Leuvenink
- Department of Surgery (Division of Experimental Surgery), University of Groningen, UMCG, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.,The NIGRAM consortium detailed in the Supplemental Acknowledgments
| |
Collapse
|
218
|
Ding H, Jiang L, Xu J, Bai F, Zhou Y, Yuan Q, Luo J, Zen K, Yang J. Inhibiting aerobic glycolysis suppresses renal interstitial fibroblast activation and renal fibrosis. Am J Physiol Renal Physiol 2017; 313:F561-F575. [PMID: 28228400 DOI: 10.1152/ajprenal.00036.2017] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/09/2017] [Accepted: 02/15/2017] [Indexed: 11/22/2022] Open
Abstract
Chronic kidney diseases generally lead to renal fibrosis. Despite great progress having been made in identifying molecular mediators of fibrosis, the mechanism that governs renal fibrosis remains unclear, and so far no effective therapeutic antifibrosis strategy is available. Here we demonstrated that a switch of metabolism from oxidative phosphorylation to aerobic glycolysis (Warburg effect) in renal fibroblasts was the primary feature of fibroblast activation during renal fibrosis and that suppressing renal fibroblast aerobic glycolysis could significantly reduce renal fibrosis. Both gene and protein assay showed that the expression of glycolysis enzymes was upregulated in mouse kidneys with unilateral ureter obstruction (UUO) surgery or in transforming growth factor-β1 (TGF-β1)-treated renal interstitial fibroblasts. Aerobic glycolysis flux, indicated by glucose uptake and lactate production, was increased in mouse kidney with UUO nephropathy or TGF-β1-treated renal interstitial fibroblasts and positively correlated with fibrosis process. In line with this, we found that increasing aerobic glycolysis can remarkably induce myofibroblast activation while aerobic glycolysis inhibitors shikonin and 2-deoxyglucose attenuate UUO-induced mouse renal fibrosis and TGF-β1-stimulated myofibroblast activation. Furthermore, mechanistic study indicated that shikonin inhibits renal aerobic glycolysis via reducing phosphorylation of pyruvate kinase type M2, a rate-limiting glycolytic enzyme associated with cell reliance on aerobic glycolysis. In conclusion, our findings demonstrate the critical role of aerobic glycolysis in renal fibrosis and support treatment with aerobic glycolysis inhibitors as a potential antifibrotic strategy.
Collapse
Affiliation(s)
- Hao Ding
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China; and
| | - Lei Jiang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China; and
| | - Jing Xu
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China; and
| | - Feng Bai
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China; and
| | - Yang Zhou
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China; and
| | - Qi Yuan
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China; and
| | - Jing Luo
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China; and
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences, Nanjing, China
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China; and
| |
Collapse
|
219
|
Chen J, Zhang H, Hu J, Gu Y, Shen Z, Xu L, Jia X, Zhang X, Ding X. Hydrogen-Rich Saline Alleviates Kidney Fibrosis Following AKI and Retains Klotho Expression. Front Pharmacol 2017; 8:499. [PMID: 28848432 PMCID: PMC5554490 DOI: 10.3389/fphar.2017.00499] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/13/2017] [Indexed: 01/16/2023] Open
Abstract
Purpose: Acute kidney injury (AKI) is a prominent risk factor for the development of chronic kidney disease (CKD). To date, the related mechanism and effective therapy have not been rigorously explored. The present study aims to investigate the reno-protection of hydrogen-rich saline (HRS) against ischemia/reperfusion (IR)-induced AKI. Methods: Adult male C57 mice were randomly allocated into three groups: Sham, IR, IR+HRS. Renal IR injury model was generated via 35 min occlusion of bilateral kidney pedicles, and then, mice were administered with different treatments intraperitoneally in various groups. After 14- or 28-day treatment, mice were perfused and the kidneys were collected following reperfusion. Many proteins were detected by western blots, including renal fibrotic proteins [a-smooth muscle actin (a-SMA), collagen I (Col I)], Klotho, the methylation of Klotho, damage-regulated autophagy modulator (Beclin-1), and microtubule-associated protein light 3-II (LC3-II). Finally, the levels of serum blood urea nitrogen (BUN) and creatinine (Cr) were measured to investigate the renal function. Results: Histological data showed that the HRS treatment significantly decreased the fibrosis in renal tissues when compared with the IR group, and both of BUN and Cr were lower in the HRS group than IR group (8.9 ± 0.6 vs. 9.9 ± 0.1 mmol/l, 51 ± 6.5 vs. 60 ± 5.8 μmol/l) (P < 0.05). The expression of fibrotic markers, a-SMA and Col I, showed a robust increase in IR injury models than the Sham group, which was consistent with the result of Trichrome staining. However, the levels of a-SMA and Col I expression were sharply decreased in the IR+HRS group (P < 0.05). IR injury also enhanced LC3-II and Beclin-1 expression, but decreased Klotho level. The Klotho level was alleviated by HRS, but LC3-II and Beclin-1 were starkly enhanced in HRS group (P < 0.05). Conclusion: HRS showed a protective effect in the prevention of renal injury and could inhibit renal fibrosis after IR injury in mice. This role of HRS might be exerted via retaining Klotho expression and activating autophagy in the kidney.
Collapse
Affiliation(s)
- Jing Chen
- Department of Nephrology, Zhongshan Hospital, Fudan UniversityShanghai, China.,Kidney and Dialysis Institute of ShanghaiShanghai, China.,Kidney and Blood Purification Laboratory of ShanghaiShanghai, China
| | - Han Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan UniversityShanghai, China.,Kidney and Dialysis Institute of ShanghaiShanghai, China.,Kidney and Blood Purification Laboratory of ShanghaiShanghai, China
| | - Jiachang Hu
- Department of Nephrology, Zhongshan Hospital, Fudan UniversityShanghai, China.,Kidney and Dialysis Institute of ShanghaiShanghai, China.,Kidney and Blood Purification Laboratory of ShanghaiShanghai, China
| | - Yulu Gu
- Kidney and Dialysis Institute of ShanghaiShanghai, China
| | - Ziyan Shen
- Department of Nephrology, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Linghan Xu
- Kidney and Dialysis Institute of ShanghaiShanghai, China
| | - Xueqi Jia
- Kidney and Dialysis Institute of ShanghaiShanghai, China
| | - Xiaoyan Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan UniversityShanghai, China.,Kidney and Dialysis Institute of ShanghaiShanghai, China.,Kidney and Blood Purification Laboratory of ShanghaiShanghai, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan UniversityShanghai, China.,Kidney and Dialysis Institute of ShanghaiShanghai, China.,Kidney and Blood Purification Laboratory of ShanghaiShanghai, China
| |
Collapse
|
220
|
Suppression of Wnt Signaling and Osteogenic Changes in Vascular Smooth Muscle Cells by Eicosapentaenoic Acid. Nutrients 2017; 9:nu9080858. [PMID: 28796175 PMCID: PMC5579651 DOI: 10.3390/nu9080858] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 01/01/2023] Open
Abstract
Vascular medial calcification is often observed in patients with arteriosclerosis. It is also associated with systolic hypertension, wide pulse pressure, and fluctuation of blood pressure, which results in cardiovascular events. Eicosapentaenoic acid (EPA) has been shown to suppress vascular calcification in previous animal experiments. We investigated the inhibitory effects of EPA on Wnt signaling, which is one of the important signaling pathways involved in vascular calcification. Intake of food containing 5% EPA resulted in upregulation of the mRNA expression of Klotho, an intrinsic inhibitor of Wnt signaling, in the kidneys of wild-type mice. Expression levels of β-catenin, an intracellular signal transducer in the Wnt signaling pathway, were increased in the aortas of Klotho mutant (kl/kl) mice compared to the levels in the aortas of wild-type mice. Wnt3a or BIO, a GSK-3 inhibitor that activates β-catenin signaling, upregulated mRNA levels of AXIN2 and LEF1, Wnt signaling marker genes, and RUNX2 and BMP4, early osteogenic genes, in human aorta smooth muscle cells. EPA suppressed the upregulation of AXIN2 and BMP4. The effect of EPA was cancelled by T0070907, a PPARγ inhibitor. The results suggested that EPA could suppress vascular calcification via the inhibition of Wnt signaling in osteogenic vascular smooth muscle cells via PPARγ activation.
Collapse
|
221
|
Poveda J, Sanz AB, Carrasco S, Ruiz-Ortega M, Cannata-Ortiz P, Sanchez-Niño MD, Ortiz A. Bcl3: a regulator of NF-κB inducible by TWEAK in acute kidney injury with anti-inflammatory and antiapoptotic properties in tubular cells. Exp Mol Med 2017; 49:e352. [PMID: 28684863 PMCID: PMC5565957 DOI: 10.1038/emm.2017.89] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/21/2016] [Accepted: 01/12/2017] [Indexed: 01/09/2023] Open
Abstract
Acute kidney injury (AKI) is characterized by tubular cell death and interstitial inflammation. TWEAK promotes experimental kidney injury and activates the transcription factor NF-κB, a key regulator of genes involved in cell survival and inflammatory response. In search of potential therapeutic targets for AKI, we compared a transcriptomics database of NF-κB-related genes from murine AKI-kidneys with a transcriptomics database of TWEAK-stimulated cultured tubular cells. Four out of twenty-four (17%) genes were significantly upregulated (false discovery rate, FDR<0.05), while nine out of twenty-four (37%) genes were significantly upregulated at FDR <0.1 in both databases. Bcl3 was the top upregulated NF-κB-related gene in experimental AKI and one of the most upregulated genes in TWEAK-stimulated tubular cells. Quantitative reverse transcription PCR (qRT-PCR), western blot and immunohistochemistry confirmed Bcl3 upregulation in both experimental conditions and localized increased Bcl3 expression to tubular cells in AKI. Transcriptomics database analysis revealed increased Bcl3 expression in numerous experimental and human kidney conditions. Furthermore, systemic TWEAK administration increased kidney Bcl3 expression. In cultured tubular cells, targeting Bcl3 by siRNA resulted in the magnification of TWEAK-induced NF-κB transcriptional activity, chemokine upregulation and Klotho downregulation, and in the sensitization to cell death induced by TWEAK/TNFα/interferon-γ. In contrast, Bcl3 overexpression decreased NF-κB transcriptional activity, inflammatory response and cell death while dampening the decrease in Klotho expression. In conclusion, Bcl3 expressed in response to TWEAK stimulation decreases TWEAK-induced inflammatory and lethal responses. Therefore, therapeutic upregulation of Bcl3 activity should be explored in kidney disease because it has advantages over chemical inhibitors of NF-κB that are known to prevent inflammatory responses but can also sensitize the cells to apoptosis.
Collapse
Affiliation(s)
- Jonay Poveda
- Unidad de Dialisis, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, Fundación Renal Iñigo Álvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| | - Ana B Sanz
- Unidad de Dialisis, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, Fundación Renal Iñigo Álvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| | - Susana Carrasco
- Unidad de Dialisis, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, Fundación Renal Iñigo Álvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| | - Marta Ruiz-Ortega
- Unidad de Dialisis, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, Fundación Renal Iñigo Álvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| | - Pablo Cannata-Ortiz
- Unidad de Dialisis, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, Fundación Renal Iñigo Álvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| | - Maria D Sanchez-Niño
- Unidad de Dialisis, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, Fundación Renal Iñigo Álvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| | - Alberto Ortiz
- Unidad de Dialisis, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, Fundación Renal Iñigo Álvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| |
Collapse
|
222
|
Gröne EF, Federico G, Nelson PJ, Arnold B, Gröne HJ. The hormetic functions of Wnt pathways in tubular injury. Pflugers Arch 2017; 469:899-906. [PMID: 28685176 PMCID: PMC5541077 DOI: 10.1007/s00424-017-2018-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/13/2017] [Accepted: 06/13/2017] [Indexed: 02/06/2023]
Abstract
Chronic tubulointerstitial damage with tubular epithelial atrophy and interstitial fibrosis is the hallmark of chronic kidney disease (CKD) and a predictor for progression of CKD.Several experiments have now provided evidence that the Wnt signaling pathways are significantly contributing to atrophy and fibrosis; in contrast, it also has been shown that the Wnt system fosters regenerative processes in acute tubular injury.We now have demonstrated that Dickkopf 3 (DKK3) is an agonist for canonical Wnt signaling in CKD and fosters chronic fibrosing inflammation of the tubulointerstitial compartment. Genetic- and antibody-mediated inhibition of DKK3 leads to a pronounced improvement of tubular differentiation and a reduction in fibrosis.In addition, the secreted glycoprotein DKK3 can be used as a non-invasive urinary marker for the extent of CKD in man.
Collapse
Affiliation(s)
- Elisabeth F Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Giuseppina Federico
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Peter J Nelson
- Clinical Biochemistry, Ludwig Maximilian University, Munich, Bavaria, Germany
| | - Bernd Arnold
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Hermann-Josef Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. h.-
| |
Collapse
|
223
|
Olauson H, Mencke R, Hillebrands JL, Larsson TE. Tissue expression and source of circulating αKlotho. Bone 2017; 100:19-35. [PMID: 28323144 DOI: 10.1016/j.bone.2017.03.043] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/16/2022]
Abstract
αKlotho (Klotho), a type I transmembrane protein and a coreceptor for Fibroblast Growth Factor-23, was initially thought to be expressed only in a limited number of tissues, most importantly the kidney, parathyroid gland and choroid plexus. Emerging data may suggest a more ubiquitous Klotho expression pattern which has prompted reevaluation of the restricted Klotho paradigm. Herein we systematically review the evidence for Klotho expression in various tissues and cell types in humans and other mammals, and discuss potential reasons behind existing conflicting data. Based on current literature and tissue expression atlases, we propose a classification of tissues into high, intermediate and low/absent Klotho expression. The functional relevance of Klotho in organs with low expression levels remain uncertain and there is currently limited data on a role for membrane-bound Klotho outside the kidney. Finally, we review the evidence for the tissue source of soluble Klotho, and conclude that the kidney is likely to be the principal source of circulating Klotho in physiology.
Collapse
Affiliation(s)
- Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Rik Mencke
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tobias E Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
224
|
Neyra JA, Hu MC. Potential application of klotho in human chronic kidney disease. Bone 2017; 100:41-49. [PMID: 28115282 PMCID: PMC5474175 DOI: 10.1016/j.bone.2017.01.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 01/13/2023]
Abstract
The extracellular domain of transmembrane alpha-Klotho (αKlotho, hereinafter simply called Klotho) is cleaved by secretases and released into the circulation as soluble Klotho. Soluble Klotho in the circulation starts to decline early in chronic kidney disease (CKD) stage 2 and urinary Klotho possibly even earlier in CKD stage 1. Therefore soluble Klotho could serve as an early and sensitive marker of kidney function decline. Moreover, preclinical animal data support Klotho deficiency is not just merely a biomarker, but a pathogenic factor for CKD progression and extrarenal CKD complications including cardiovascular disease and disturbed mineral metabolism. Prevention of Klotho decline, re-activation of endogenous Klotho production or supplementation of exogenous Klotho are all associated with attenuation of renal fibrosis, retardation of CKD progression, improvement of mineral metabolism, amelioration of cardiomyopathy, and alleviation of vascular calcification in CKD. Therefore Klotho is not only a diagnostic and/or prognostic marker for CKD, but the treatment of Klotho deficiency may be a promising strategy to prevent, retard, and decrease the burden of comorbidity in CKD.
Collapse
Affiliation(s)
- Javier A Neyra
- Department of Internal Medicine, University of Texas Southwestern Medical Center, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, USA
| | - Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, USA.
| |
Collapse
|
225
|
Ó hAinmhire E, Humphreys BD. Fibrotic Changes Mediating Acute Kidney Injury to Chronic Kidney Disease Transition. Nephron Clin Pract 2017; 137:264-267. [PMID: 28595180 DOI: 10.1159/000474960] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/02/2017] [Indexed: 12/24/2022] Open
Abstract
End-stage renal disease (ESRD) is common, costly, and it results from progressive chronic kidney disease (CKD). ESRD claims many lives every year. It is increasingly recognized that episodes of acute kidney injury (AKI) predispose to the future development of CKD and ESRD. While our understanding of the pathophysiology of the AKI to CKD transition is improving, there are no validated therapeutic strategies to prevent this transition. In this review, we summarize the recent progress made in defining the cellular and molecular events underlying the AKI to CKD transition and highlight potential therapeutic targets and strategies to reduce the incidence of CKD following AKI.
Collapse
Affiliation(s)
- Eoghainín Ó hAinmhire
- Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
226
|
Drew DA, Katz R, Kritchevsky S, Ix J, Shlipak M, Gutiérrez OM, Newman A, Hoofnagle A, Fried L, Semba RD, Sarnak M. Association between Soluble Klotho and Change in Kidney Function: The Health Aging and Body Composition Study. J Am Soc Nephrol 2017; 28:1859-1866. [PMID: 28104822 PMCID: PMC5461794 DOI: 10.1681/asn.2016080828] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/10/2016] [Indexed: 11/03/2022] Open
Abstract
CKD appears to be a condition of soluble klotho deficiency. Despite known associations between low soluble klotho levels and conditions that promote kidney damage, such as oxidative stress and fibrosis, little information exists regarding the longitudinal association between soluble klotho levels and change in kidney function. We assayed serum soluble α-klotho in 2496 participants within the Health Aging and Body Composition study, a cohort of older adults. The associations between soluble klotho levels and decline in kidney function (relative decline: eGFR decline ≥30%; absolute decline: eGFR decline >3 ml/min per year) and incident CKD (incident eGFR <60 ml/min per 1.73 m2 and >1 ml/min per year decline) were evaluated. We adjusted models for demographics, baseline eGFR, urine albumin-to-creatinine ratio, comorbidity, and measures of mineral metabolism. Among participants, the mean (SD) age was 75 (3) years, 52% were women, and 38% were black. Median (25th, 75th percentiles) klotho level was 630 (477, 817) pg/ml. In fully adjusted models, each two-fold higher level of klotho associated with lower odds of decline in kidney function (odds ratio, 0.78 [95% confidence interval, 0.66 to 0.93] for 30% decline in eGFR, and 0.85 [95% confidence interval, 0.73 to 0.98] for >3 ml/min per year decline in eGFR), but not of incident CKD (incident rate ratio, 0.90 [95% confidence interval, 0.78 to 1.04]). Overall, a higher soluble klotho level independently associated with a lower risk of decline in kidney function. Future studies should attempt to replicate these results in other cohorts and evaluate the underlying mechanism.
Collapse
Affiliation(s)
- David A Drew
- Department of Medicine, Division of Nephrology, Tufts Medical Center, Boston, Massachusetts;
| | - Ronit Katz
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, Washington
| | - Stephen Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Joachim Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA and Nephrology Section, Veterans Affairs San Diego Healthcare System, La Jolla, California
| | - Michael Shlipak
- Kidney Health Research Collaborative, San Francisco VA Medical Center and University of California, San Francisco, San Francisco, California; Departments of
| | - Orlando M Gutiérrez
- Medicine and
- Epidemiology, University of Alabama School of Medicine, Birmingham, Alabama
| | - Anne Newman
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Andy Hoofnagle
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, Washington
| | - Linda Fried
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, Washington
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, Washington
- Renal Section, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania and
| | - Richard D Semba
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Mark Sarnak
- Department of Medicine, Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
227
|
Susnik N, Sen P, Melk A, Schmitt R. Aging, Cellular Senescence, and Kidney Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0143-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
228
|
Muñoz-Castañeda JR, Herencia C, Pendón-Ruiz de Mier MV, Rodriguez-Ortiz ME, Diaz-Tocados JM, Vergara N, Martínez-Moreno JM, Salmerón MD, Richards WG, Felsenfeld A, Kuro-O M, Almadén Y, Rodríguez M. Differential regulation of renal Klotho and FGFR1 in normal and uremic rats. FASEB J 2017; 31:3858-3867. [PMID: 28515153 DOI: 10.1096/fj.201700006r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/24/2017] [Indexed: 01/03/2023]
Abstract
In renal failure, hyperphosphatemia occurs despite a marked elevation in serum fibroblast growth factor (FGF)-23. Abnormal regulation of the FGFR1-Klotho receptor complex may cause a resistance to the phosphaturic action of FGF23. The purpose of the present study was to investigate the regulation of renal Klotho and FGF receptor (FEFR)-1 in healthy and uremic rats induced by 5/6 nephrectomy. In normal rats, the infusion of rat recombinant FGF23 enhanced phosphaturia and increased renal FGFR1 expression; however, Klotho expression was reduced. Uremic rats on a high-phosphate (HP) diet presented hyperphosphatemia with marked elevation of FGF23 and an increased fractional excretion of phosphate (P) that was associated with a marked reduction of Klotho expression and an increase in FGFR1. After neutralization of FGF23 by anti-FGF23 administration, phosphaturia was still abundant, Klotho expression remained low, and the FGFR1 level was reduced. These results suggest that the expression of renal Klotho is modulated by phosphaturia, whereas the FGFR1 expression is regulated by FGF23. Calcitriol (CTR) administration prevented a decrease in renal Klotho expression. In HEK293 cells HP produced nuclear translocation of β-catenin, together with a reduction in Klotho. Wnt/β-catenin inhibition with Dkk-1 prevented the P-induced down-regulation of Klotho. The addition of CTR to HP medium was able to recover Klotho expression. In summary, high FGF23 levels increase FGFR1, whereas phosphaturia decreases Klotho expression through the activation of Wnt/β-catenin pathway.-Muñoz-Castañeda, J. R., Herencia, C., Pendón-Ruiz de Mier, M. V., Rodriguez-Ortiz, M. E., Diaz-Tocados, J. M., Vergara, N., Martínez-Moreno, J. M., Salmerón, M. D., Richards, W. G., Felsenfeld, A., Kuro-O, M., Almadén, Y., Rodríguez, M. Differential regulation of renal Klotho and FGFR1 in normal and uremic rats.
Collapse
Affiliation(s)
- Juan R Muñoz-Castañeda
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Unidad de Gestión Clínica (UGC) Nefrología, Hospital Universitario Reina Sofía, Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Red Nacional de Investigación en Nefrología (REDinREN), Instituto de Salud Carlos III, Córdoba, Spain
| | - Carmen Herencia
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Maria Victoria Pendón-Ruiz de Mier
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain
| | | | - Juan M Diaz-Tocados
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Noemi Vergara
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Julio M Martínez-Moreno
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Maria Dolores Salmerón
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain
| | | | - Arnold Felsenfeld
- Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System and the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yolanda Almadén
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Unidad de Gestión Clínica (UGC) Medicina Interna, Hospital Universitario Reina Sofía, Córdoba, Spain.,Centro de Investigación Biomédica en Red Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Córdoba, Spain
| | - Mariano Rodríguez
- Instituto Maimónides para la Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain; .,Unidad de Gestión Clínica (UGC) Nefrología, Hospital Universitario Reina Sofía, Córdoba, Spain.,Departamento de Medicina, Universidad de Córdoba, Córdoba, Spain.,Red Nacional de Investigación en Nefrología (REDinREN), Instituto de Salud Carlos III, Córdoba, Spain
| |
Collapse
|
229
|
DNA hypermethylation of sFRP5 contributes to indoxyl sulfate-induced renal fibrosis. J Mol Med (Berl) 2017; 95:601-613. [PMID: 28508124 DOI: 10.1007/s00109-017-1538-0] [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: 11/06/2016] [Revised: 03/24/2017] [Accepted: 04/27/2017] [Indexed: 12/21/2022]
Abstract
Renal fibrosis is the most common outcome of chronic kidney disease (CKD), while the pathogenesis of renal fibrosis is not fully understood. In this study, we first showed that the progress of renal fibrosis was positively related to serum levels of indoxyl sulfate, a typical protein-bound toxin, and that there was a close correlation between serum indoxyl sulfate levels and β-catenin expression in the kidneys (r = 0.908, p < 0.001) of CKD patients. We then demonstrated that intraperitoneal injections of indoxyl sulfate (100 mg/kg/day) for 4 weeks in uninephrectomized mice explicitly induced renal fibrosis, which was accompanied by a significant activation of Wnt/β-catenin signaling. In vitro investigations in human renal tubular HK-2 cells revealed that indoxyl sulfate exhibited a potent ability to induce Wnt/β-catenin activation through the downregulation of sFRP5, a gene that codes for an extracellular antagonist of Wnt signaling, by increasing the DNA methylation level of its promoter CpG islands. The increased expression of DNA methyltransferases following the activation of ROS/ERK1/2 signaling was responsible for the DNA hypermethylation of sFRP5 induced by indoxyl sulfate. Conversely, treatment with 5-aza-2'-deoxycytidine, an inhibitor of DNA methyltransferases, significantly reduced indoxyl sulfate-induced sFRP5 downregulation and Wnt/β-catenin activation. In vivo, intraperitoneal injections of recombinant sFRP5 protein or 5-aza-2'-deoxycytidine substantially alleviated renal fibrosis in indoxyl sulfate-treated uninephrectomized mice. Our results suggest that indoxyl sulfate promotes renal fibrosis through the induction of DNA hypermethylation of sFRP5, and thereafter the activation of Wnt/β-catenin signaling. These findings provide new insights into the pathogenesis of renal fibrosis in CKD patients. KEY MESSAGES IS induces renal fibrosis by increasing ß-catenin expression in CKD mice. IS-induced Wnt signaling activation is due to sFRP5 hypermethylation in HK-2 cells. ROS/ERK1/2 signaling activation is involved in IS-induced sFRP5 hypermethylation. sFRP5 upregulation attenuates IS-induced renal fibrosis by inhibiting Wnt signaling.
Collapse
|
230
|
Mencke R, Hillebrands JL. The role of the anti-ageing protein Klotho in vascular physiology and pathophysiology. Ageing Res Rev 2017; 35:124-146. [PMID: 27693241 DOI: 10.1016/j.arr.2016.09.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/23/2016] [Indexed: 02/08/2023]
Abstract
Klotho is an anti-ageing protein that functions in many pathways that govern ageing, like regulation of phosphate homeostasis, insulin signaling, and Wnt signaling. Klotho expression levels and levels in blood decline during ageing. The vascular phenotype of Klotho deficiency features medial calcification, intima hyperplasia, endothelial dysfunction, arterial stiffening, hypertension, and impaired angiogenesis and vasculogenesis, with characteristics similar to aged human arteries. Klotho-deficient phenotypes can be prevented and rescued by Klotho gene expression or protein supplementation. High phosphate levels are likely to be directly pathogenic and are a prerequisite for medial calcification, but more important determinants are pathways that regulate cellular senescence, suggesting that deficiency of Klotho renders cells susceptible to phosphate toxicity. Overexpression of Klotho is shown to ameliorate medial calcification, endothelial dysfunction, and hypertension. Endogenous vascular Klotho expression is a controversial subject and, currently, no compelling evidence exists that supports the existence of vascular membrane-bound Klotho expression, as expressed in kidney. In vitro, Klotho has been shown to decrease oxidative stress and apoptosis in both SMCs and ECs, to reduce SMC calcification, to maintain the contractile SMC phenotype, and to prevent μ-calpain overactivation in ECs. Klotho has many protective effects with regard to the vasculature and constitutes a very promising therapeutic target. The purpose of this review is to explore the etiology of the vascular phenotype of Klotho deficiency and the therapeutic potential of Klotho in vascular disease.
Collapse
|
231
|
Lin X, Zha Y, Zeng XZ, Dong R, Wang QH, Wang DT. Role of the Wnt/β-catenin signaling pathway in inducing apoptosis and renal fibrosis in 5/6-nephrectomized rats. Mol Med Rep 2017; 15:3575-3582. [PMID: 28440442 PMCID: PMC5436235 DOI: 10.3892/mmr.2017.6461] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 02/14/2017] [Indexed: 12/22/2022] Open
Abstract
Renal fibrosis is the final common pathway of all progressive renal disease. Excessive and chronic activation of the Wnt/β-catenin signaling pathway results in chronic kidney disease (CKD) progression. To mimic CKD, the present study used 5/6-nephrectomized rats, and alterations in kidney histology, expression of β-catenin and renal cell apoptosis were assessed. In addition, mesangial cells were cultured in vitro and transfected with β-catenin siRNA to evaluate the effect of blocking Wnt/β-catenin signaling on cell apoptosis and the expression of markers of renal fibrosis. The results demonstrated that CKD rat kidney tissues exhibited moderate renal fibrosis and significantly increased expression levels of β-catenin and apoptosis associated proteins compared with sham-operated rats. In vitro, silencing of β-catenin by siRNA attenuated tumor necrosis factor-α-induced apoptosis and decreased mRNA expression levels of various markers of fibrosis, including fibronectin, transforming growth factor-β, and collagen I, III and IV. In conclusion, inhibition of Wnt/β-catenin signaling by β-catenin silencing attenuated apoptosis and expression of fibrosis-associated markers in renal cells. The present study suggested that the Wnt/β-catenin signaling pathway may serve as a potential treatment strategy for renal fibrotic disorders.
Collapse
Affiliation(s)
- Xin Lin
- Department of Nephrology, People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Yan Zha
- Department of Nephrology, People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Xiang-Zhen Zeng
- Department of Nephrology, Loudi Central Hospital, Loudi, Hunan 417000, P.R. China
| | - Rong Dong
- Department of Nephrology, People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Qing-Hua Wang
- Department of Nephrology, Loudi Central Hospital, Loudi, Hunan 417000, P.R. China
| | - Dong-Tao Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| |
Collapse
|
232
|
Li Y, Hu F, Xue M, Jia YJ, Zheng ZJ, Wang L, Guan MP, Xue YM. Klotho down-regulates Egr-1 by inhibiting TGF-β1/Smad3 signaling in high glucose treated human mesangial cells. Biochem Biophys Res Commun 2017; 487:216-222. [PMID: 28411025 DOI: 10.1016/j.bbrc.2017.04.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 01/26/2023]
Abstract
Diabetic kidney disease (DKD) has become the leading cause of end-stage renal disease worldwide and is associated with glomerular mesangial cell (MC) proliferation and excessive extracellular matrix (ECM) production. Klotho can attenuate renal fibrosis in part by inhibiting TGF-β1/Smad3 signaling in DKD. Early growth response factor 1 (Egr-1) has been shown to play a key role in renal fibrosis in part by facilitating the formation of a positive feedback loop involving TGF-β1. However, whether Klotho down-regulates Egr-1 by inhibiting TGF-β1/Smad3 signaling in DKD is unclear. In the present study, we assessed human MCs that were incubated under high-glucose conditions to mimic diabetes. Then, we transfected the cells with Klotho plasmid or siRNA to overexpress or knock down Klotho gene and protein expression. Klotho, Egr-1, fibronectin (FN), collagen type I (Col I), Smad3 and phosphorylated Smad3 (p-Smad3) gene and protein expression levels were determined by RT-qPCR and western blotting respectively. High glucose time-dependently down-regulated Klotho mRNA and protein expression in cultured human MCs. pcDNA3.1-Klotho transfection-mediated Klotho overexpression down-regulated Egr-1, FN and Col I expression and the p-Smad3/Smad3 ratio in human MCs. Conversely, siRNA-mediated Klotho silencing up-regulated Egr-1, FN, and Col I expression and the p-Smad3/Smad3 ratio. Moreover, the effects of si-Klotho on Egr-1 expression were abolished by the TGF-β1 inhibitor SB-431542. Klotho overexpression can prevent mesangial ECM production in high-glucose-treated human MCs, an effect that has been partially attributed to Egr-1 down-regulation facilitated by TGF-β1/Smad3 signaling inhibition.
Collapse
Affiliation(s)
- Yang Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Geriatrics, Zhu Jiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fang Hu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Meng Xue
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Endocrinology and Metabolism, Shenzhen People's Hospital, Second Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Yi-Jie Jia
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zong-Ji Zheng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ling Wang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mei-Ping Guan
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yao-Ming Xue
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
233
|
Li Q, Vo HT, Wang J, Fox-Quick S, Dobrunz LE, King GD. Klotho regulates CA1 hippocampal synaptic plasticity. Neuroscience 2017; 347:123-133. [PMID: 28215989 PMCID: PMC5392240 DOI: 10.1016/j.neuroscience.2017.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 01/07/2023]
Abstract
Global klotho overexpression extends lifespan while global klotho-deficiency shortens it. As well, klotho protein manipulations inversely regulate cognitive function. Mice without klotho develop rapid onset cognitive impairment before they are 2months old. Meanwhile, adult mice overexpressing klotho show enhanced cognitive function, particularly in hippocampal-dependent tasks. The cognitive enhancing effects of klotho extend to humans with a klotho polymorphism that increases circulating klotho and executive function. To affect cognitive function, klotho could act in or on the synapse to modulate synaptic transmission or plasticity. However, it is not yet known if klotho is located at synapses, and little is known about its effects on synaptic function. To test this, we fractionated hippocampi and detected klotho expression in both pre and post-synaptic compartments. We find that loss of klotho enhances both pre and post-synaptic measures of CA1 hippocampal synaptic plasticity at 5weeks of age. However, a rapid loss of synaptic enhancement occurs such that by 7weeks, when mice are cognitively impaired, there is no difference from wild-type controls. Klotho overexpressing mice show no early life effects on synaptic plasticity, but decreased CA1 hippocampal long-term potentiation was measured at 6months of age. Together these data suggest that klotho affects cognition, at least in part, by regulating hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Qin Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jing Wang
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stephanie Fox-Quick
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lynn E Dobrunz
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
234
|
Wei SY, Wang YX, Zhang QF, Zhao SL, Diao TT, Li JS, Qi WR, He YX, Guo XY, Zhang MZ, Chen JY, Wang XT, Wei QJ, Wang Y, Li B. Multiple Mechanisms are Involved in Salt-Sensitive Hypertension-Induced Renal Injury and Interstitial Fibrosis. Sci Rep 2017; 7:45952. [PMID: 28383024 PMCID: PMC5382679 DOI: 10.1038/srep45952] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 03/08/2017] [Indexed: 12/20/2022] Open
Abstract
Salt-sensitive hypertension (SSHT) leads to kidney interstitial fibrosis. However, the potential mechanisms leading to renal fibrosis have not been well investigated. In present study, Dahl salt-sensitive (DS) rats were divided into three groups: normal salt diet (DSN), high salt diet (DSH) and high salt diet treated with hydrochlorothiazide (HCTZ) (DSH + HCTZ). A significant increase in systolic blood pressure (SBP) was observed 3 weeks after initiating the high salt diet, and marked histological alterations were observed in DSH rats. DSH rats showed obvious podocyte injury, peritubular capillary (PTC) loss, macrophage infiltration, and changes in apoptosis and cell proliferation. Moreover, Wnt/β-catenin signaling was significantly activated in DSH rats. However, HCTZ administration attenuated these changes with decreased SBP. In addition, increased renal and urinary Wnt4 expression was detected with time in DSH rats and was closely correlated with histopathological alterations. Furthermore, these alterations were also confirmed by clinical study. In conclusion, the present study provides novel insight into the mechanisms related to PTC loss, macrophage infiltration and Wnt/β-catenin signaling in SSHT-induced renal injury and fibrosis. Therefore, multi-target therapeutic strategies may be the most effective in preventing these pathological processes. Moreover, urinary Wnt4 may be a noninvasive biomarker for monitoring renal injury after hypertension.
Collapse
Affiliation(s)
- Shi-Yao Wei
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yu-Xiao Wang
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Qing-Fang Zhang
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Shi-Lei Zhao
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Tian-Tian Diao
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Jian-Si Li
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Wen-Rui Qi
- Financial Mathematics, Beijing Normal University-Hong Kong Baptist University United International College Zhuhai, People’s Republic of China
| | - Yi-Xin He
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xin-Yu Guo
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Man-Zhu Zhang
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Jian-Yu Chen
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xiao-Ting Wang
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Qiu-Ju Wei
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yu Wang
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Bing Li
- Department of Nephrology, Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
235
|
González N, Prieto I, del Puerto-Nevado L, Portal-Nuñez S, Ardura JA, Corton M, Fernández-Fernández B, Aguilera O, Gomez-Guerrero C, Mas S, Moreno JA, Ruiz-Ortega M, Sanz AB, Sanchez-Niño MD, Rojo F, Vivanco F, Esbrit P, Ayuso C, Alvarez-Llamas G, Egido J, García-Foncillas J, Ortiz A. 2017 update on the relationship between diabetes and colorectal cancer: epidemiology, potential molecular mechanisms and therapeutic implications. Oncotarget 2017; 8:18456-18485. [PMID: 28060743 PMCID: PMC5392343 DOI: 10.18632/oncotarget.14472] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/26/2016] [Indexed: 02/06/2023] Open
Abstract
Worldwide deaths from diabetes mellitus (DM) and colorectal cancer increased by 90% and 57%, respectively, over the past 20 years. The risk of colorectal cancer was estimated to be 27% higher in patients with type 2 DM than in non-diabetic controls. However, there are potential confounders, information from lower income countries is scarce, across the globe there is no correlation between DM prevalence and colorectal cancer incidence and the association has evolved over time, suggesting the impact of additional environmental factors. The clinical relevance of these associations depends on understanding the mechanism involved. Although evidence is limited, insulin use has been associated with increased and metformin with decreased incidence of colorectal cancer. In addition, colorectal cancer shares some cellular and molecular pathways with diabetes target organ damage, exemplified by diabetic kidney disease. These include epithelial cell injury, activation of inflammation and Wnt/β-catenin pathways and iron homeostasis defects, among others. Indeed, some drugs have undergone clinical trials for both cancer and diabetic kidney disease. Genome-wide association studies have identified diabetes-associated genes (e.g. TCF7L2) that may also contribute to colorectal cancer. We review the epidemiological evidence, potential pathophysiological mechanisms and therapeutic implications of the association between DM and colorectal cancer. Further studies should clarify the worldwide association between DM and colorectal cancer, strengthen the biological plausibility of a cause-and-effect relationship through characterization of the molecular pathways involved, search for specific molecular signatures of colorectal cancer under diabetic conditions, and eventually explore DM-specific strategies to prevent or treat colorectal cancer.
Collapse
Affiliation(s)
- Nieves González
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz-UAM, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Isabel Prieto
- Radiation Oncology, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Laura del Puerto-Nevado
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Sergio Portal-Nuñez
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Juan Antonio Ardura
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Marta Corton
- Genetics, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Oscar Aguilera
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Sebastián Mas
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | | | - Ana Belen Sanz
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
- REDINREN, Madrid, Spain
| | | | - Federico Rojo
- Pathology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Pedro Esbrit
- Bone and Mineral Metabolism laboratory, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Carmen Ayuso
- Genetics, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | | | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundacion Jimenez Diaz-UAM, Spanish Biomedical Research Network in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Alberto Ortiz
- Nephrology, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
- REDINREN, Madrid, Spain
| | | |
Collapse
|
236
|
Yin S, Zhang Q, Yang J, Lin W, Li Y, Chen F, Cao W. TGFβ-incurred epigenetic aberrations of miRNA and DNA methyltransferase suppress Klotho and potentiate renal fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1207-1216. [PMID: 28285987 DOI: 10.1016/j.bbamcr.2017.03.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 12/31/2022]
Abstract
Renal fibrosis is a common pathological feature of chronic kidney diseases (CKD) and its development and progression are significantly affected by epigenetic modifications such as aberrant miRNA and DNA methylation. Klotho is an anti-aging and anti-fibrotic protein and its early decline after renal injury is reportedly associated with aberrant DNA methylation. However, the key upstream pathological mediators and the molecular cascade leading to epigenetic Klotho suppression are not exclusively established. Here we investigate the epigenetic mechanism of Klotho deficiency and its functional relevance in renal fibrogenesis. Fibrotic kidneys induced by unilateral ureteral occlusion (UUO) displayed marked Klotho suppression and the promoter hypermethylation. These abnormalities were likely due to deregulated transforming growth factor-beta (TGFβ) since TGFβ alone caused the similar epigenetic aberrations in cultured renal cells and TGFβ blockade prevented the alterations in UUO kidney. Further investigation revealed that TGFβ enhanced DNA methyltransferase (DNMT) 1 and DNMT3a via inhibiting miR-152 and miR-30a in both renal cells and fibrotic kidneys. Accordingly the blockade of either TGFβ signaling or DNMT1/3a activities significantly recovered the Klotho loss and attenuated pro-fibrotic protein expression and renal fibrosis. Moreover, Klotho knockdown by RNA interferences abolished the anti-fibrotic effects of DNMT inhibition in both TGFβ-treated renal cell and UUO kidney, indicating that TGFβ-mediated miR-152/30a inhibitions, DNMT1/3a aberrations and subsequent Klotho loss constitute a critical regulatory loop that eliminates Klotho's anti-fibrotic activities and potentiates renal fibrogenesis. Thus, our study elaborates a novel epigenetic cascade of renal fibrogenesis and reveals the potential therapeutic targets for treating the renal fibrosis-associated kidney diseases.
Collapse
Affiliation(s)
- Shasha Yin
- Nanjing University School of Medicine, Jiangsu, Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Qin Zhang
- Nanjing University School of Medicine, Jiangsu, Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jun Yang
- Nanjing University School of Medicine, Jiangsu, Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Wenjun Lin
- Nanjing University School of Medicine, Jiangsu, Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Yanning Li
- Nanjing University School of Medicine, Jiangsu, Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Fang Chen
- Nanjing University School of Medicine, Jiangsu, Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Wangsen Cao
- Nanjing University School of Medicine, Jiangsu, Key Laboratory of Molecular Medicine, Nanjing 210093, China.
| |
Collapse
|
237
|
Yang T, Xu C. Physiology and Pathophysiology of the Intrarenal Renin-Angiotensin System: An Update. J Am Soc Nephrol 2017; 28:1040-1049. [PMID: 28255001 DOI: 10.1681/asn.2016070734] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The renin-angiotensin system (RAS) has a pivotal role in the maintenance of extracellular volume homeostasis and blood pressure through complex mechanisms. Apart from the well known systemic RAS, occurrence of a local RAS has been documented in multiple tissues, including the kidney. A large body of recent evidence from pharmacologic and genetic studies, particularly those using various transgenic approaches to manipulate intrarenal levels of RAS components, has established the important role of intrarenal RAS in hypertension. Recent studies have also begun to unravel the molecular mechanisms that govern intrarenal RAS activity. This local system is under the control of complex regulatory networks consisting of positive regulators of (pro)renin receptor, Wnt/β-catenin signaling, and PGE2/PGE2 receptor EP4 subtype, and negative regulators of Klotho, vitamin D receptor, and liver X receptors. This review highlights recent advances in defining the regulation and function of intrarenal RAS as a unique entity separate from systemic angiotensin II generation.
Collapse
Affiliation(s)
- Tianxin Yang
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and .,Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| | - Chuanming Xu
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China
| |
Collapse
|
238
|
Soluble klotho binds monosialoganglioside to regulate membrane microdomains and growth factor signaling. Proc Natl Acad Sci U S A 2017; 114:752-757. [PMID: 28069944 DOI: 10.1073/pnas.1620301114] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Soluble klotho, the shed ectodomain of the antiaging membrane protein α-klotho, is a pleiotropic endocrine/paracrine factor with no known receptors and poorly understood mechanism of action. Soluble klotho down-regulates growth factor-driven PI3K signaling, contributing to extension of lifespan, cardioprotection, and tumor inhibition. Here we show that soluble klotho binds membrane lipid rafts. Klotho binding to rafts alters lipid organization, decreases membrane's propensity to form large ordered domains for endocytosis, and down-regulates raft-dependent PI3K/Akt signaling. We identify α2-3-sialyllactose present in the glycan of monosialogangliosides as targets of soluble klotho. α2-3-Sialyllactose is a common motif of glycans. To explain why klotho preferentially targets lipid rafts we show that clustering of gangliosides in lipid rafts is important. In vivo, raft-dependent PI3K signaling is up-regulated in klotho-deficient mouse hearts vs. wild-type hearts. Our results identify ganglioside-enriched lipid rafts to be receptors that mediate soluble klotho regulation of PI3K signaling. Targeting sialic acids may be a general mechanism for pleiotropic actions of soluble klotho.
Collapse
|
239
|
Kim JH, Xie J, Hwang KH, Wu YL, Oliver N, Eom M, Park KS, Barrezueta N, Kong ID, Fracasso RP, Huang CL, Cha SK. Klotho May Ameliorate Proteinuria by Targeting TRPC6 Channels in Podocytes. J Am Soc Nephrol 2017; 28:140-151. [PMID: 27151926 PMCID: PMC5198269 DOI: 10.1681/asn.2015080888] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 03/18/2016] [Indexed: 12/30/2022] Open
Abstract
Klotho is a type-1 membrane protein predominantly produced in the kidney, the extracellular domain of which is secreted into the systemic circulation. Membranous and secreted Klotho protect organs, including the kidney, but whether and how Klotho directly protects the glomerular filter is unknown. Here, we report that secreted Klotho suppressed transient receptor potential channel 6 (TRPC6)-mediated Ca2+ influx in cultured mouse podocytes by inhibiting phosphoinositide 3-kinase-dependent exocytosis of the channel. Furthermore, soluble Klotho reduced ATP-stimulated actin cytoskeletal remodeling and transepithelial albumin leakage in these cells. Overexpression of TRPC6 by gene delivery in mice induced albuminuria, and exogenous administration of Klotho ameliorated the albuminuria. Notably, immunofluorescence and in situ hybridization revealed Klotho expression in podocytes of mouse and human kidney. Heterozygous Klotho-deficient CKD mice had aggravated albuminuria compared with that in wild-type CKD mice with a similar degree of hypertension and reduced clearance function. Finally, disrupting the integrity of glomerular filter by saline infusion-mediated extracellular fluid volume expansion increased urinary Klotho excretion. These results reveal a potential novel function of Klotho in protecting the glomerular filter, and may offer a new therapeutic strategy for treatment of proteinuria.
Collapse
Affiliation(s)
- Ji-Hee Kim
- Departments of Physiology and Global Medical Science
| | - Jian Xie
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kyu-Hee Hwang
- Departments of Physiology and Global Medical Science
| | - Yueh-Lin Wu
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan; and
| | - Noelynn Oliver
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut
| | | | - Kyu-Sang Park
- Departments of Physiology and Global Medical Science
- Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Nestor Barrezueta
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut
| | - In-Deok Kong
- Departments of Physiology and Global Medical Science
- Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - R Paul Fracasso
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut
| | - Chou-Long Huang
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Seung-Kuy Cha
- Departments of Physiology and Global Medical Science,
- Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| |
Collapse
|
240
|
Berchtold L, Ponte B, Moll S, Hadaya K, Seyde O, Bachtler M, Vallée JP, Martin PY, Pasch A, de Seigneux S. Phosphocalcic Markers and Calcification Propensity for Assessment of Interstitial Fibrosis and Vascular Lesions in Kidney Allograft Recipients. PLoS One 2016; 11:e0167929. [PMID: 28036331 PMCID: PMC5201285 DOI: 10.1371/journal.pone.0167929] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/22/2016] [Indexed: 11/18/2022] Open
Abstract
Renal interstitial fibrosis and arterial lesions predict loss of function in chronic kidney disease. Noninvasive estimation of interstitial fibrosis and vascular lesions is currently not available. The aim of the study was to determine whether phosphocalcic markers are associated with, and can predict, renal chronic histological changes. We included 129 kidney allograft recipients with an available transplant biopsy in a retrospective study. We analyzed the associations and predictive values of phosphocalcic markers and serum calcification propensity (T50) for chronic histological changes (interstitial fibrosis and vascular lesions). PTH, T50 and vitamin D levels were independently associated to interstitial fibrosis. PTH elevation was associated with increasing interstitial fibrosis severity (r = 0.29, p = 0.001), while T50 and vitamin D were protective (r = -0.20, p = 0.025 and r = -0.23, p = 0.009 respectively). On the contrary, fibroblast growth factor 23 (FGF23) and Klotho correlated only modestly with interstitial fibrosis (p = 0.045) whereas calcium and phosphate did not. PTH, vitamin D and T50 were predictors of extensive fibrosis (AUC: 0.73, 0.72 and 0.68 respectively), but did not add to renal function prediction. PTH, FGF23 and T50 were modestly predictive of low fibrosis (AUC: 0.63, 0.63 and 0.61) but did not add to renal function prediction. T50 decreased with increasing arterial lesions (r = -0.21, p = 0.038). The discriminative performance of T50 in predicting significant vascular lesions was modest (AUC 0.61). In summary, we demonstrated that PTH, vitamin D and T50 are associated to interstitial fibrosis and vascular lesions in kidney allograft recipients independently of renal function. Despite these associations, mineral metabolism indices do not show superiority or additive value to fibrosis prediction by eGFR and proteinuria in kidney allograft recipients, except for vascular lesions where T50 could be of relevance.
Collapse
Affiliation(s)
- Lena Berchtold
- Service of Internal Medicine, Department of Internal Medicine, University Hospital of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Internal Medicine Specialities, University Hospital of Geneva, Geneva, Switzerland
| | - Belen Ponte
- Service of Nephrology, Department of Internal Medicine Specialities, University Hospital of Geneva, Geneva, Switzerland
| | - Solange Moll
- Institute of Clinical Pathology, Departement of Clinical Pathology, University Hospital of Geneva, Geneva, Switzerland
| | - Karine Hadaya
- Service of Nephrology, Department of Internal Medicine Specialities, University Hospital of Geneva, Geneva, Switzerland
| | - Olivia Seyde
- Institute of Clinical Pathology, Departement of Clinical Pathology, University Hospital of Geneva, Geneva, Switzerland
| | - Matthias Bachtler
- Service of Clinical Research, Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Jean-Paul Vallée
- Service of Radiology, Department of Radiology and Medical Informatics, University Hospital of Geneva, Geneva, Switzerland
| | - Pierre-Yves Martin
- Service of Nephrology, Department of Internal Medicine Specialities, University Hospital of Geneva, Geneva, Switzerland
| | - Andreas Pasch
- Service of Clinical Research, Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Sophie de Seigneux
- Service of Nephrology, Department of Internal Medicine Specialities, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
241
|
Wu YL, Xie J, An SW, Oliver N, Barrezueta NX, Lin MH, Birnbaumer L, Huang CL. Inhibition of TRPC6 channels ameliorates renal fibrosis and contributes to renal protection by soluble klotho. Kidney Int 2016; 91:830-841. [PMID: 27979597 DOI: 10.1016/j.kint.2016.09.039] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 09/15/2016] [Accepted: 09/22/2016] [Indexed: 12/19/2022]
Abstract
Fibrosis is an exaggerated form of tissue repair that occurs with serious damage or repetitive injury and ultimately leads to organ failure due to the excessive scarring. Increased calcium ion entry through the TRPC6 channel has been associated with the pathogenesis of heart and glomerular diseases, but its role in renal interstitial fibrosis is unknown. We studied this by deletion of Trpc6 in mice and found it decreased unilateral ureteral obstruction-induced interstitial fibrosis and blunted increased mRNA expression of fibrosis-related genes in the ureteral obstructed kidney relative to that in the kidney of wild-type mice. Administration of BTP2, a pyrazol derivative known to inhibit function of several TRPC channels, also ameliorated obstruction-induced renal fibrosis and gene expression in wild-type mice. BTP2 inhibited carbachol-activated TRPC3 and TRPC6 channel activities in HEK293 cells. Ureteral obstruction caused over a 10-fold increase in mRNA expression for TRPC3 as well as TRPC6 in the kidneys of obstructed relative to the sham-operated mice. The magnitude of protection against obstruction-induced fibrosis in Trpc3 and Trpc6 double knockout mice was not different from that in Trpc6 knockout mice. Klotho, a membrane and soluble protein predominantly produced in the kidney, is known to confer protection against renal fibrosis. Administration of soluble klotho significantly reduced obstruction-induced renal fibrosis in wild-type mice, but not in Trpc6 knockout mice, indicating that klotho and TRPC6 inhibition act in the same pathway to protect against obstruction-induced renal fibrosis. Thus klotho and TRPC6 may be pharmacologic targets for treating renal fibrosis.
Collapse
Affiliation(s)
- Yueh-Lin Wu
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jian Xie
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sung-Wan An
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Noelynn Oliver
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Nestor X Barrezueta
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Mei-Hsiang Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Lutz Birnbaumer
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA; Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina
| | - Chou-Long Huang
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
242
|
Xiao Z, Riccardi D, Velazquez HA, Chin AL, Yates CR, Carrick JD, Smith JC, Baudry J, Quarles LD. A computationally identified compound antagonizes excess FGF-23 signaling in renal tubules and a mouse model of hypophosphatemia. Sci Signal 2016; 9:ra113. [PMID: 27879395 PMCID: PMC6544179 DOI: 10.1126/scisignal.aaf5034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fibroblast growth factor-23 (FGF-23) interacts with a binary receptor complex composed of α-Klotho (α-KL) and FGF receptors (FGFRs) to regulate phosphate and vitamin D metabolism in the kidney. Excess FGF-23 production, which causes hypophosphatemia, is genetically inherited or occurs with chronic kidney disease. Among other symptoms, hypophosphatemia causes vitamin D deficiency and the bone-softening disorder rickets. Current therapeutics that target the receptor complex have limited utility clinically. Using a computationally driven, structure-based, ensemble docking and virtual high-throughput screening approach, we identified four novel compounds predicted to selectively inhibit FGF-23-induced activation of the FGFR/α-KL complex. Additional modeling and functional analysis found that Zinc13407541 bound to FGF-23 and disrupted its interaction with the FGFR1/α-KL complex; experiments in a heterologous cell expression system showed that Zinc13407541 selectivity inhibited α-KL-dependent FGF-23 signaling. Zinc13407541 also inhibited FGF-23 signaling in isolated renal tubules ex vivo and partially reversed the hypophosphatemic effects of excess FGF-23 in a mouse model. These chemical probes provide a platform to develop lead compounds to treat disorders caused by excess FGF-23.
Collapse
Affiliation(s)
- Zhousheng Xiao
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38165, USA
| | - Demian Riccardi
- Department of Chemistry, Earlham College, 801 National Road West, Richmond, IN 47374, USA
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Hector A Velazquez
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
- Department of Chemistry, Tennessee Technological University, 55 University Drive, Cookeville, TN 38501, USA
| | - Ai L Chin
- University of Tennessee/Oak Ridge National Laboratory Center for Molecular Biophysics, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN 37830, USA
| | - Charles R Yates
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jesse D Carrick
- University of Tennessee/Oak Ridge National Laboratory Center for Molecular Biophysics, Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN 37830, USA
| | - Jeremy C Smith
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
- Department of Chemistry, Tennessee Technological University, 55 University Drive, Cookeville, TN 38501, USA
| | - Jerome Baudry
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
- Department of Chemistry, Tennessee Technological University, 55 University Drive, Cookeville, TN 38501, USA
| | - L Darryl Quarles
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38165, USA.
| |
Collapse
|
243
|
Abstract
Individuals age >65 years old are the fastest expanding population demographic throughout the developed world. Consequently, more aged patients than before are receiving diagnoses of impaired renal function and nephrosclerosis-age-associated histologic changes in the kidneys. Recent studies have shown that the aged kidney undergoes a range of structural changes and has altered transcriptomic, hemodynamic, and physiologic behavior at rest and in response to renal insults. These changes impair the ability of the kidney to withstand and recover from injury, contributing to the high susceptibility of the aged population to AKI and their increased propensity to develop subsequent progressive CKD. In this review, we examine these features of the aged kidney and explore the various validated and putative pathways contributing to the changes observed with aging in both experimental animal models and humans. We also discuss the potential for additional study to increase understanding of the aged kidney and lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Eoin D O'Sullivan
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom;
| | - Jeremy Hughes
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; and
| | - David A Ferenbach
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom.,MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; and.,Renal and.,Biomedical Engineering Divisions, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
244
|
Zhou L, Liu Y. Wnt/β-catenin signaling and renin-angiotensin system in chronic kidney disease. Curr Opin Nephrol Hypertens 2016; 25:100-6. [PMID: 26808707 DOI: 10.1097/mnh.0000000000000205] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Intrarenal activation of the renin-angiotensin system (RAS) plays an essential role in the pathogenesis of hypertension and chronic kidney diseases (CKD). However, how RAS genes are regulated in vivo was poorly understood until recently. This review focuses on recent findings of the transcriptional regulation of RAS components, as well as their implication in developing novel strategies to treat the patients with CKD. RECENT FINDINGS Bioinformatics analyses have uncovered the presence of putative binding sites for T-cell factor/β-catenin in the promoter region of all RAS genes. Both in-vitro and in-vivo studies confirm that Wnt/β-catenin is the master upstream regulator that controls the expression of all RAS components tested, such as angiotensinogen, renin, angiotensin converting enzyme and the angiotensin II type I receptor in the kidney. Targeted inhibition of Wnt/β-catenin, by either small molecule ICG-001 or endogenous Wnt antagonist Klotho, represses RAS activation and ameliorates proteinuria and kidney injury. Blockade of Wnt/β-catenin signaling also normalizes blood pressure in a mouse model of CKD. SUMMARY These recent studies identify Wnt/β-catenin as the master regulator that controls multiple RAS genes, and suggest that targeting this upstream signaling could be an effective strategy for the treatment of patients with hypertension and CKD.
Collapse
Affiliation(s)
- Lili Zhou
- aState Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China bDepartment of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
245
|
Keap1 hypomorphism protects against ischemic and obstructive kidney disease. Sci Rep 2016; 6:36185. [PMID: 27804998 PMCID: PMC5090361 DOI: 10.1038/srep36185] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 10/10/2016] [Indexed: 12/26/2022] Open
Abstract
The Keap1/Nrf2 pathway is a master regulator of antioxidant, anti-inflammatory, and other cytoprotective mechanisms important in protection from kidney disease. For the first time in kidney disease, we describe the use of Keap1 hypomorphic mice, which possess Nrf2 hyperactivation. We exposed these mice and wild type controls to ischemia/reperfusion injury (IRI). The initial tubular injury at 24 hours post-IRI appeared to be unaffected, with the only observed difference being a decrease in inflammatory cytokine expression in the hypomorphs. However, we noted significant improvement in serum creatinine in the hypomorphs at 3 and 10 days after injury, and renal fibrosis was dramatically attenuated at the late timepoint. Assessment of Nrf2-regulated targets (GSTM1, GSTP1, NQO1) revealed higher expression in the hypomorphs at baseline. While injury tended to suppress these genes in wild-type mice, the suppression was attenuated or reversed in Keap1 hypomorphs, suggesting that protection in these mice was mediated by increased Nrf2 transcriptional activity. To assess the generalizability of our findings, we subjected the hypomorphs to unilateral ureteral obstruction (UUO) and again found significant protection and increased expression of Nrf2 targets. Overall, these results support the conclusion that the Nrf2 pathway is protective in a variety of kidney diseases.
Collapse
|
246
|
Ruiz-Andres O, Sanchez-Niño MD, Moreno JA, Ruiz-Ortega M, Ramos AM, Sanz AB, Ortiz A. Downregulation of kidney protective factors by inflammation: role of transcription factors and epigenetic mechanisms. Am J Physiol Renal Physiol 2016; 311:F1329-F1340. [PMID: 27760772 DOI: 10.1152/ajprenal.00487.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 12/22/2022] Open
Abstract
Chronic kidney disease (CKD) is associated to an increased risk of death, CKD progression, and acute kidney injury (AKI) even from early stages, when glomerular filtration rate (GFR) is preserved. The link between early CKD and these risks is unclear, since there is no accumulation of uremic toxins. However, pathological albuminuria and kidney inflammation are frequent features of early CKD, and the production of kidney protective factors may be decreased. Indeed, Klotho expression is already decreased in CKD category G1 (normal GFR). Klotho has anti-aging and nephroprotective properties, and decreased Klotho levels may contribute to increase the risk of death, CKD progression, and AKI. In this review, we discuss the downregulation by mediators of inflammation of molecules with systemic and/or renal local protective functions, exemplified by Klotho and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), a transcription factor that promotes mitochondrial biogenesis. Cytokines such as TWEAK, TNF-α, or transforming growth factor -β1 produced locally during kidney injury or released from inflammatory sites at other organs may decrease kidney expression of Klotho and PGC-1α or lead to suboptimal recruitment of these nephroprotective proteins. Transcription factors (e.g., Smad3 and NF-κB) and epigenetic mechanisms (e.g., histone acetylation or methylation) contribute to downregulate the expression of Klotho and/or PGC-1α, while histone crotonylation promotes PGC-1α expression. NF-κBiz facilitates the repressive effect of NF-κB on Klotho expression. A detailed understanding of these mediators may contribute to the development of novel therapeutic approaches to prevent CKD progression and its negative impact on mortality and AKI.
Collapse
Affiliation(s)
- Olga Ruiz-Andres
- IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid.,REDINREN, Madrid, Spain; and
| | - Maria Dolores Sanchez-Niño
- IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid.,REDINREN, Madrid, Spain; and
| | - Juan Antonio Moreno
- IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid
| | - Marta Ruiz-Ortega
- IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid.,REDINREN, Madrid, Spain; and
| | - Adrian Mario Ramos
- IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid.,REDINREN, Madrid, Spain; and
| | - Ana Belen Sanz
- IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid.,REDINREN, Madrid, Spain; and
| | - Alberto Ortiz
- IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid; .,REDINREN, Madrid, Spain; and.,Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| |
Collapse
|
247
|
Zhang Q, Yin S, Liu L, Liu Z, Cao W. Rhein reversal of DNA hypermethylation-associated Klotho suppression ameliorates renal fibrosis in mice. Sci Rep 2016; 6:34597. [PMID: 27703201 PMCID: PMC5050540 DOI: 10.1038/srep34597] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/15/2016] [Indexed: 11/15/2022] Open
Abstract
Renal fibrosis is the hallmark of chronic kidney diseases (CKD) and its development and progression are significantly affected by epigenetic modifications. Rhein, a plant-derived anthraquinone, displays strong anti-fibrosis properties, but its protective mode of action remains incompletely understood. Here we explore the mechanism of Rhein anti-renal fibrosis by investigating its regulation of Klotho, a known renal anti-fibrotic protein whose suppression after renal injury reportedly involves aberrant DNA methylation. We report that Rhein is an impressive up-regulator of Klotho and it markedly reversed Klotho down-regulation in unilateral ureteral occlusion-induced fibrotic kidney. Further examinations revealed that Klotho loss in fibrotic kidney is associated with Klotho promoter hypermethylation due to aberrant methyltransferase 1 and 3a expressions. However, Rhein significantly corrected all these epigenetic alterations and subsequently alleviated pro-fibrotic protein expression and renal fibrosis, whereas Klotho knockdown via RNA interferences largely abrogated the anti-renal fibrotic effects of Rhein, suggesting that Rhein epigenetic reversal of Klotho loss represents a critical mode of action that confers Rhein’s anti- renal fibrotic functions. Altogether our studies uncover a novel hypomethylating character of Rhein in preventing Klotho loss and renal fibrosis, and demonstrate the efficacy of Klotho-targeted epigenetic intervention in potential treatment of renal fibrosis-associated kidney diseases.
Collapse
Affiliation(s)
- Qin Zhang
- Division of Nephrology, Jinling Hospital, Southern Medical University, Nanjing, 210016, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210016, China
| | - Shasha Yin
- The Key lab of Jiangsu molecular Medicine, Nanjing University School of Medicine, Nanjing, 210093, China
| | - Lin Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210016, China
| | - Zhihong Liu
- Division of Nephrology, Jinling Hospital, Southern Medical University, Nanjing, 210016, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210016, China
| | - Wangsen Cao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210016, China.,The Key lab of Jiangsu molecular Medicine, Nanjing University School of Medicine, Nanjing, 210093, China
| |
Collapse
|
248
|
Zhou D, Tian Y, Sun L, Zhou L, Xiao L, Tan RJ, Tian J, Fu H, Hou FF, Liu Y. Matrix Metalloproteinase-7 Is a Urinary Biomarker and Pathogenic Mediator of Kidney Fibrosis. J Am Soc Nephrol 2016; 28:598-611. [PMID: 27624489 DOI: 10.1681/asn.2016030354] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/07/2016] [Indexed: 12/19/2022] Open
Abstract
Matrix metalloproteinase-7 (MMP-7), a secreted zinc- and calcium-dependent endopeptidase, is a transcriptional target of canonical Wnt/β-catenin signaling. Because Wnt/β-catenin is activated in diseased kidney, we hypothesized that urinary MMP-7 level may be used as a noninvasive surrogate biomarker for fibrotic lesions. To test this hypothesis, we conducted a cross-sectional study, measuring urinary MMP-7 levels in a cohort of 102 patients with CKD. Compared with normal subjects, patients with various kidney disorders had markedly elevated urinary levels of MMP-7. Furthermore, urinary MMP-7 levels closely correlated with renal fibrosis scores in patients. In mice, knockout of MMP-7 ameliorated the fibrotic lesions and expression of matrix genes induced by obstructive injury. Genetic ablation of MMP-7 also preserved E-cadherin protein expression and substantially reduced the expression of total and dephosphorylated β-catenin and the de novo expression of vimentin and fibroblast-specific protein 1 in renal tubules of obstructed kidneys. In vitro, MMP-7 proteolytically degraded E-cadherin in proximal tubular cells, leading to β-catenin liberation and nuclear translocation and induction of β-catenin target genes by a mechanism independent of Wnt ligands. Finally, pharmacologic inhibition of MMP-7 immediately after obstructive injury reduced renal fibrosis in vivo These results suggest that MMP-7 not only can serve as a noninvasive biomarker but also is an important pathogenic mediator of kidney fibrosis.
Collapse
Affiliation(s)
| | - Yuan Tian
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ling Sun
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liangxiang Xiao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Roderick J Tan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Jianwei Tian
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- Department of Pathology and .,State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
249
|
Falke LL, Kinashi H, Dendooven A, Broekhuizen R, Stoop R, Joles JA, Nguyen TQ, Goldschmeding R. Age-dependent shifts in renal response to injury relate to altered BMP6/CTGF expression and signaling. Am J Physiol Renal Physiol 2016; 311:F926-F934. [PMID: 27558559 DOI: 10.1152/ajprenal.00324.2016] [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: 05/31/2016] [Accepted: 08/17/2016] [Indexed: 02/07/2023] Open
Abstract
Age is associated with an increased prevalence of chronic kidney disease (CKD), which, through progressive tissue damage and fibrosis, ultimately leads to loss of kidney function. Although much effort is put into studying CKD development experimentally, age has rarely been taken into account. Therefore, we investigated the effect of age on the development of renal tissue damage and fibrosis in a mouse model of obstructive nephropathy (i.e., unilateral ureter obstruction; UUO). We observed that after 14 days, obstructed kidneys of old mice had more tubulointerstitial atrophic damage but less fibrosis than those of young mice. This was associated with reduced connective tissue growth factor (CTGF), and higher bone morphogenetic protein 6 (BMP6) expression and pSMAD1/5/8 signaling, while transforming growth factor-β expression and transcriptional activity were no different in obstructed kidneys of old and young mice. In vitro, CTGF bound to and inhibited BMP6 activity. In summary, our data suggest that in obstructive nephropathy atrophy increases and fibrosis decreases with age and that this relates to increased BMP signaling, most likely due to higher BMP6 and lower CTGF expression.
Collapse
Affiliation(s)
- Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hiroshi Kinashi
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Nephrology and Renal Replacement Therapy, Nagoya University, Nagoya, Japan
| | - Amelie Dendooven
- Department of Pathology, University Medical Center, Antwerp, Belgium
| | - Roel Broekhuizen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Reinout Stoop
- Department of Metabolic Health Research, TNO, Leiden, The Netherlands; and
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands;
| |
Collapse
|
250
|
The Hippo-Salvador signaling pathway regulates renal tubulointerstitial fibrosis. Sci Rep 2016; 6:31931. [PMID: 27550469 PMCID: PMC4994041 DOI: 10.1038/srep31931] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/28/2016] [Indexed: 12/22/2022] Open
Abstract
Renal tubulointerstitial fibrosis (TIF) is the final pathway of various renal injuries that result in chronic kidney disease. The mammalian Hippo-Salvador signaling pathway has been implicated in the regulation of cell proliferation, cell death, tissue regeneration, and tumorigenesis. Here, we report that the Hippo-Salvador pathway plays a role in disease development in patients with TIF and in a mouse model of TIF. Mice with tubular epithelial cell (TEC)-specific deletions of Sav1 (Salvador homolog 1) exhibited aggravated renal TIF, enhanced epithelial-mesenchymal transition-like phenotypic changes, apoptosis, and proliferation after unilateral ureteral obstruction (UUO). Moreover, Sav1 depletion in TECs increased transforming growth factor (TGF)-β and activated β-catenin expression after UUO, which likely accounts for the abovementioned enhanced TEC fibrotic phenotype. In addition, TAZ (transcriptional coactivator with PDZ-binding motif), a major downstream effector of the Hippo pathway, was significantly activated in Sav1-knockout mice in vivo. An in vitro study showed that TAZ directly regulates TGF-β and TGF-β receptor II expression. Collectively, our data indicate that the Hippo-Salvador pathway plays a role in the pathogenesis of TIF and that regulating this pathway may be a therapeutic strategy for reducing TIF.
Collapse
|