1
|
Vanichapol T, Gonzalez A, Delgado R, Brewer M, Clouthier KA, Menshikh A, Snyder WE, Rahman T, Sander V, Yang H, Davidson A, de Caestecker M. Partial repair causes permanent defects in papillary structure and function after reversal of urinary obstruction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612436. [PMID: 39314319 PMCID: PMC11419032 DOI: 10.1101/2024.09.11.612436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Urinary obstruction causes injury to the renal papilla and leads to defects in the ability to concentrate urine which predisposes to progressive kidney injury. However, the regenerative capacity of the papilla after reversal of obstruction is poorly understood. To address this, we developed a mouse model of reversible urinary obstruction which is characterized by extensive papillary injury, followed by a robust regeneration response and complete histological recovery over a 3- month period. However, these mice have a pronounced defect in urinary concentrating capacity. We now show that this is due to permanent changes in the composition, organization, and transcriptional signatures of epithelial, endothelial, and interstitial cell lineages in the papilla. There are persistent inflammatory responses that are also seen in patients with renal stone disease but are associated with cell-specific adaptive responses to the increasingly hypoxic environment of the papilla after reversal of obstruction. Taken together, our analysis of a new model of reversible urinary obstruction reveals that partial repair leads to permanent changes in the structure and function of all of the major cellular compartments in the papilla that include both shared and distinct responses to different types of renal papillary injury in humans and mice. Summary Partial repair after reversal of urinary obstruction leads to permanent changes in structure and function of all major cellular compartments in the renal papilla.
Collapse
|
2
|
Lathan R. Exploring unconventional targets in myofibroblast transdifferentiation outside classical TGF- β signaling in renal fibrosis. Front Physiol 2024; 15:1296504. [PMID: 38808357 PMCID: PMC11130449 DOI: 10.3389/fphys.2024.1296504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
We propose that the key initiators of renal fibrosis are myofibroblasts which originate from four predominant sources-fibroblasts, pericytes, endothelial cells and macrophages. Increased accumulation of renal interstitial myofibroblasts correlates with an increase in collagen, fibrillar proteins, and fibrosis severity. The canonical TGF-β pathway, signaling via Smad proteins, is the central molecular hub that initiates these cellular transformations. However, directly targeting these classical pathway molecules has proven challenging due their integral roles in metabolic process, and/or non-sustainable effects involving compensatory cross-talk with TGF-β. This review explores recently discovered alternative molecular targets that drive transdifferentiation into myofibroblasts. Discovering targets outside of the classical TGF-β/Smad pathway is crucial for advancing antifibrotic therapies, and strategically targeting the development of myofibroblasts offers a promising approach to control excessive extracellular matrix deposition and impede fibrosis progression.
Collapse
Affiliation(s)
- Rashida Lathan
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
3
|
Huang B, Han R, Tan H, Zhu W, Li Y, Jiang F, Xie C, Ren Z, Shi R. Scutellarin ameliorates diabetic nephropathy via TGF-β1 signaling pathway. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:25. [PMID: 38656633 PMCID: PMC11043297 DOI: 10.1007/s13659-024-00446-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024]
Abstract
Breviscapine, a natural flavonoid mixture derived from the traditional Chinese herb Erigeron breviscapus (Vant.) Hand-Mazz, has demonstrated a promising potential in improving diabetic nephropathy (DN). However, the specific active constituent(s) responsible for its therapeutic effects and the underlying pharmacological mechanisms remain unclear. In this study, we aimed to investigate the impact of scutellarin, a constituent of breviscapine, on streptozotocin-induced diabetic nephropathy and elucidate its pharmacological mechanism(s). Our findings demonstrate that scutellarin effectively ameliorates various features of DN in vivo, including proteinuria, glomerular expansion, mesangial matrix accumulation, renal fibrosis, and podocyte injury. Mechanistically, scutellarin appears to exert its beneficial effects through modulation of the transforming growth factor-β1 (TGF-β1) signaling pathway, as well as its interaction with the extracellular signal-regulated kinase (Erk) and Wnt/β-catenin pathways.
Collapse
Affiliation(s)
- Bangrui Huang
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource (Ministry of Education)Yunnan Provincial Center for Research and Development of Natural Products, School of Pharmacy, Yunnan University, Kunming, 650500, People's Republic of China
| | - Rui Han
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Hong Tan
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Wenzhuo Zhu
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Yang Li
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Fakun Jiang
- Key Laboratory of Medicinal Chemistry for Natural Resource (Ministry of Education)Yunnan Provincial Center for Research and Development of Natural Products, School of Pharmacy, Yunnan University, Kunming, 650500, People's Republic of China
| | - Chun Xie
- Key Laboratory of Medicinal Chemistry for Natural Resource (Ministry of Education)Yunnan Provincial Center for Research and Development of Natural Products, School of Pharmacy, Yunnan University, Kunming, 650500, People's Republic of China
| | - Zundan Ren
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China
| | - Rou Shi
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, People's Republic of China.
| |
Collapse
|
4
|
Yang X, Zhao H, Li R, Chen Y, Xu Z, Shang Z. Stromal thrombospondin 1 suppresses angiogenesis in oral submucous fibrosis. Int J Oral Sci 2024; 16:17. [PMID: 38403794 PMCID: PMC10894862 DOI: 10.1038/s41368-024-00286-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/14/2023] [Accepted: 01/21/2024] [Indexed: 02/27/2024] Open
Abstract
A decline in mucosal vascularity is a histological hallmark of oral submucous fibrosis (OSF), a premalignant disease that is largely induced by betel quid chewing. However, the lack of available models has challenged studies of angiogenesis in OSF. Here, we found that the expression of thrombospondin 1 (THBS1), an endogenous angiostatic protein, was elevated in the stroma of tissues with OSF. Using a fibroblast-attached organoid (FAO) model, the overexpression of THBS1 in OSF was stably recapitulated in vitro. In the FAO model, treatment with arecoline, a major pathogenic component in areca nuts, enhanced the secretion of transforming growth factor (TGF)-β1 by epithelial cells, which then promoted the expression of THBS1 in fibroblasts. Furthermore, human umbilical vein endothelial cells (HUVECs) were incorporated into the FAO to mimic the vascularized component. Overexpression of THBS1 in fibroblasts drastically suppressed the sprouting ability of endothelial cells in vascularized FAOs (vFAOs). Consistently, treatment with arecoline reduced the expression of CD31 in vFAOs, and this effect was attenuated when the endothelial cells were preincubated with neutralizing antibody of CD36, a receptor of THBS1. Finally, in an arecoline-induced rat OSF model, THBS1 inhibition alleviated collagen deposition and the decline in vascularity in vivo. Overall, we exploited an assembled organoid model to study OSF pathogenesis and provide a rationale for targeting THBS1.
Collapse
Affiliation(s)
- Xiao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hui Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Rui Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Xu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
5
|
Hammoud S, Ivanova A, Osaki Y, Funk S, Yang H, Viquez O, Delgado R, Lu D, Phillips Mignemi M, Tonello J, Colon S, Lantier L, Wasserman DH, Humphreys BD, Koenitzer J, Kern J, de Caestecker M, Finkel T, Fogo A, Messias N, Lodhi IJ, Gewin LS. Tubular CPT1A deletion minimally affects aging and chronic kidney injury. JCI Insight 2024; 9:e171961. [PMID: 38516886 PMCID: PMC11063933 DOI: 10.1172/jci.insight.171961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Kidney tubules use fatty acid oxidation (FAO) to support their high energetic requirements. Carnitine palmitoyltransferase 1A (CPT1A) is the rate-limiting enzyme for FAO, and it is necessary to transport long-chain fatty acids into mitochondria. To define the role of tubular CPT1A in aging and injury, we generated mice with tubule-specific deletion of Cpt1a (Cpt1aCKO mice), and the mice were either aged for 2 years or injured by aristolochic acid or unilateral ureteral obstruction. Surprisingly, Cpt1aCKO mice had no significant differences in kidney function or fibrosis compared with wild-type mice after aging or chronic injury. Primary tubule cells from aged Cpt1aCKO mice had a modest decrease in palmitate oxidation but retained the ability to metabolize long-chain fatty acids. Very-long-chain fatty acids, exclusively oxidized by peroxisomes, were reduced in kidneys lacking tubular CPT1A, consistent with increased peroxisomal activity. Single-nuclear RNA-Seq showed significantly increased expression of peroxisomal FAO enzymes in proximal tubules of mice lacking tubular CPT1A. These data suggest that peroxisomal FAO may compensate in the absence of CPT1A, and future genetic studies are needed to confirm the role of peroxisomal β-oxidation when mitochondrial FAO is impaired.
Collapse
Affiliation(s)
- Safaa Hammoud
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Alla Ivanova
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Yosuke Osaki
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Steven Funk
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Haichun Yang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Olga Viquez
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Rachel Delgado
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Dongliang Lu
- Division of Endocrinology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Jane Tonello
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Selene Colon
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Louise Lantier
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - David H. Wasserman
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Benjamin D. Humphreys
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jeffrey Koenitzer
- Division of Pulmonary Critical Care Medicine, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Justin Kern
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Toren Finkel
- Aging Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Agnes Fogo
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nidia Messias
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Leslie S. Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Medicine, Veterans Affairs Hospital, St. Louis, Missouri, USA
| |
Collapse
|
6
|
Gebert M, Heimbucher J, Gsell VK, Keimer K, Dillinger AE, Tamm ER. Induced Attenuation of Scleral TGF-β Signaling in Mutant Mice Increases Susceptibility to IOP-Induced Optic Nerve Damage. Invest Ophthalmol Vis Sci 2024; 65:48. [PMID: 38294803 PMCID: PMC10839816 DOI: 10.1167/iovs.65.1.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
Purpose Axonal optic nerve (ON) damage in glaucoma is characteristically associated with increased amounts of active transforming growth factor-beta 2 (TGF-β2) in the ON head. Here we investigated the functional role of scleral TGF-β signaling in glaucoma. Methods A deficiency of Tgfbr2, which encodes for TGF-β receptor type II (TGF-βRII), the essential receptor for canonical TGF-β signaling, was induced in fibroblasts (including those of the sclera) of mutant mice. To this end, 5-week-old mice were treated with tamoxifen eye drops. Experimental glaucoma was induced in 8-week-old mice using a magnetic microbead (MB) model. After 6 weeks of high intraocular pressure (IOP), the ON axons and their somata in the retina were labeled by paraphenylenediamine (PPD) and RNA-binding protein with multiple splicing (RBPMS) immunohistochemistry, respectively, and quantified. Results Tamoxifen treatment resulted in a significant decrease of TGF-βRII and its mRNA in the sclera. After 6 weeks of high IOP, reduced numbers of PPD-stained ON axons were seen in MB-injected eyes in comparison with not-injected contralateral eyes. Moreover, MB injection also led to a decrease of retinal ganglion cell (RGC) somata as seen in RBPMS-stained retinal wholemounts. Axon loss and RGC loss were significantly higher in mice with a fibroblast specific deficiency of TGF-βRII in comparison with control animals. Conclusions We conclude that the ablation of scleral TGF-β signaling increases the susceptibility to IOP-induced ON damage. Scleral TGF-β signaling in mutant mice appears to be beneficial for ON axon survival in experimentally induced glaucoma.
Collapse
Affiliation(s)
- Magdalena Gebert
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Johanna Heimbucher
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Valentina K. Gsell
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Kristof Keimer
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Andrea E. Dillinger
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Ernst R. Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
7
|
Cao S, Pan Y, Terker AS, Arroyo Ornelas JP, Wang Y, Tang J, Niu A, Kar SA, Jiang M, Luo W, Dong X, Fan X, Wang S, Wilson MH, Fogo A, Zhang MZ, Harris RC. Epidermal growth factor receptor activation is essential for kidney fibrosis development. Nat Commun 2023; 14:7357. [PMID: 37963889 PMCID: PMC10645887 DOI: 10.1038/s41467-023-43226-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/03/2023] [Indexed: 11/16/2023] Open
Abstract
Fibrosis is the progressive accumulation of excess extracellular matrix and can cause organ failure. Fibrosis can affect nearly every organ including kidney and there is no specific treatment currently. Although Epidermal Growth Factor Receptor (EGFR) signaling pathway has been implicated in development of kidney fibrosis, underlying mechanisms by which EGFR itself mediates kidney fibrosis have not been elucidated. We find that EGFR expression increases in interstitial myofibroblasts in human and mouse fibrotic kidneys. Selective EGFR deletion in the fibroblast/pericyte population inhibits interstitial fibrosis in response to unilateral ureteral obstruction, ischemia or nephrotoxins. In vivo and in vitro studies and single-nucleus RNA sequencing analysis demonstrate that EGFR activation does not induce myofibroblast transformation but is necessary for the initial pericyte/fibroblast migration and proliferation prior to subsequent myofibroblast transformation by TGF-ß or other profibrotic factors. These findings may also provide insight into development of fibrosis in other organs and in other conditions.
Collapse
Affiliation(s)
- Shirong Cao
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Yu Pan
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andrew S Terker
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Juan Pablo Arroyo Ornelas
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Yinqiu Wang
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Jiaqi Tang
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Aolei Niu
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Sarah Abu Kar
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Mengdi Jiang
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Wentian Luo
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Xinyu Dong
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Xiaofeng Fan
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Suwan Wang
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Matthew H Wilson
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
- Veterans Affairs, Nashville, TN, USA
| | - Agnes Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA.
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA.
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Nashville, TN, USA.
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA.
- Veterans Affairs, Nashville, TN, USA.
| |
Collapse
|
8
|
Kayhan M, Vouillamoz J, Rodriguez DG, Bugarski M, Mitamura Y, Gschwend J, Schneider C, Hall A, Legouis D, Akdis CA, Peter L, Rehrauer H, Gewin L, Wenger RH, Khodo SN. Intrinsic TGF-β signaling attenuates proximal tubule mitochondrial injury and inflammation in chronic kidney disease. Nat Commun 2023; 14:3236. [PMID: 37270534 PMCID: PMC10239443 DOI: 10.1038/s41467-023-39050-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/26/2023] [Indexed: 06/05/2023] Open
Abstract
Excessive TGF-β signaling and mitochondrial dysfunction fuel chronic kidney disease (CKD) progression. However, inhibiting TGF-β failed to impede CKD in humans. The proximal tubule (PT), the most vulnerable renal segment, is packed with giant mitochondria and injured PT is pivotal in CKD progression. How TGF-β signaling affects PT mitochondria in CKD remained unknown. Here, we combine spatial transcriptomics and bulk RNAseq with biochemical analyses to depict the role of TGF-β signaling on PT mitochondrial homeostasis and tubulo-interstitial interactions in CKD. Male mice carrying specific deletion of Tgfbr2 in the PT have increased mitochondrial injury and exacerbated Th1 immune response in the aristolochic acid model of CKD, partly, through impaired complex I expression and mitochondrial quality control associated with a metabolic rewiring toward aerobic glycolysis in the PT cells. Injured S3T2 PT cells are identified as the main mediators of the maladaptive macrophage/dendritic cell activation in the absence of Tgfbr2. snRNAseq database analyses confirm decreased TGF-β receptors and a metabolic deregulation in the PT of CKD patients. This study describes the role of TGF-β signaling in PT mitochondrial homeostasis and inflammation in CKD, suggesting potential therapeutic targets that might be used to mitigate CKD progression.
Collapse
Affiliation(s)
- Merve Kayhan
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | | | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Zurich, Switzerland
| | - Julia Gschwend
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Andrew Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - David Legouis
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, Hospital and University of Geneva, Geneva, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Zurich, Switzerland
| | - Leary Peter
- Functional Genomics Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Leslie Gewin
- Department of Internal Medicine, Division of Nephrology, Washington University, St. Louis, USA
- Department of Medicine, St. Louis Veterans Affairs, St. Louis, USA
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
9
|
Widjaja AA, Viswanathan S, Shekeran SG, Adami E, Lim WW, Chothani S, Tan J, Goh JWT, Chen HM, Lim SY, Boustany-Kari CM, Hawkins J, Petretto E, Hübner N, Schafer S, Coffman TM, Cook SA. Targeting endogenous kidney regeneration using anti-IL11 therapy in acute and chronic models of kidney disease. Nat Commun 2022; 13:7497. [PMID: 36470928 PMCID: PMC9723120 DOI: 10.1038/s41467-022-35306-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/27/2022] [Indexed: 12/12/2022] Open
Abstract
The kidney has large regenerative capacity, but this is compromised when kidney damage is excessive and renal tubular epithelial cells (TECs) undergo SNAI1-driven growth arrest. Here we investigate the role of IL11 in TECs, kidney injury and renal repair. IL11 stimulation of TECs induces ERK- and p90RSK-mediated GSK3β inactivation, SNAI1 upregulation and pro-inflammatory gene expression. Mice with acute kidney injury upregulate IL11 in TECs leading to SNAI1 expression and kidney dysfunction, which is not seen in Il11 deleted mice or in mice administered a neutralizing IL11 antibody in either preemptive or treatment modes. In acute kidney injury, anti-TGFβ reduces renal fibrosis but exacerbates inflammation and tubule damage whereas anti-IL11 reduces all pathologies. Mice with TEC-specific deletion of Il11ra1 have reduced pathogenic signaling and are protected from renal injury-induced inflammation, fibrosis, and failure. In a model of chronic kidney disease, anti-IL11 therapy promotes TEC proliferation and parenchymal regeneration, reverses fibroinflammation and restores renal mass and function. These data highlight IL11-induced mesenchymal transition of injured TECs as an important renal pathology and suggest IL11 as a therapeutic target for restoring stalled endogenous regeneration in the diseased kidney.
Collapse
Affiliation(s)
- Anissa A Widjaja
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.
| | - Sivakumar Viswanathan
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Shamini G Shekeran
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Eleonora Adami
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.,Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
| | - Wei-Wen Lim
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Sonia Chothani
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Jessie Tan
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Joyce Wei Ting Goh
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Hui Mei Chen
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Sze Yun Lim
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | | | - Julie Hawkins
- Boehringer Ingelheim, CardioMetabolic Disease Research, Berlin, Germany
| | - Enrico Petretto
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany.,Charité-Universitätsmedizin, 10117, Berlin, Germany
| | - Sebastian Schafer
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Thomas M Coffman
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Stuart A Cook
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore. .,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore. .,MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London, UK.
| |
Collapse
|
10
|
Sang Y, Tsuji K, Fukushima K, Takahashi K, Kitamura S, Wada J. Semaporin3A-inhibitor ameliorates renal fibrosis through the regulation of JNK signaling. Am J Physiol Renal Physiol 2021; 321:F740-F756. [PMID: 34747196 DOI: 10.1152/ajprenal.00234.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal fibrosis is the common pathological pathway in progressive renal diseases. In the study, we analyzed the roles of Semaphorin 3A (SEMA3A) on renal fibrosis and the effect of SEMA3A-inhibitor (SEMA3A-I) using unilateral ureteral obstruction (UUO) mouse model. The expression of SEMA3A in the proximal tubulus and neuropilin-1 (NRP1), a recepor of SEMA3A, in fibloblast and tubular cells were increased in the UUO kidneys. The increased expression of myofibroblast marker tenascin-C and fibronection as well as renal fibrosis were increased in UUO kidneys, all of which were ameliorated by SEMA3A-I. In addition, c-Jun N-terminal kinase (JNK) signaling pathway known as the target of SEMA3A signaling, was activated in proximal tubular cells and fibroblast cells after UUO surgery while SEMA3A-I significantly attenuated the activation. In vitro, treatments with SEMA3A as well as transforming growth factor-β1 (TGF-β1) in human proximal tubular cells lost epithelial cell characters while SEMA3A-I significantly ameliorated this transformation. JNK inhibitor, SP600125, partially reversed SEMA3A and TGF-β1-induced cell transformation, indicating that JNK signaling is involved in SEMA3A-induced renal fibrosis. In addition, the treatment with SEMA3A in fibroblast cells activated the expression of tenascin-C, collagen type I and fibronection, indicating that SEMA3A may accelerate renal fibrosis through the activation of fibroblast cells. The analysis of human data revealed the positive correlation between urinary SEMA3A and urinary N-acetyl-β-D-glucosaminidase, indicating the association between SEMA3A and tubular injury. In conclusion, SEMA3A signaling is involved in renal fibrosis through JNK signaling pathway and SEMA3A-I might be the therapeutic option for protecting from renal fibrosis.
Collapse
Affiliation(s)
- Yizhen Sang
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University, Okayama, Japan
| | - Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University, Okayama, Japan
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University, Okayama, Japan
| | - Kensaku Takahashi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University, Okayama, Japan
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University, Okayama, Japan
| |
Collapse
|
11
|
Targeting of Smad7 in Mesenchymal Cells Does Not Exacerbate Fibrosis During Experimental Chronic Pancreatitis. Pancreas 2021; 50:1427-1434. [PMID: 35041343 DOI: 10.1097/mpa.0000000000001951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Transforming growth factor-β (TGF-β)-mediated accumulation of extracellular matrix proteins such as collagen I is a common feature of fibrosis. Pancreatic stellate cells play an integral role in the pathogenesis of pancreatitis, and their profibrotic ability is mainly mediated by TGF-β signaling. To specifically address the role of fibrogenic cells in experimental pancreatic fibrosis, we deleted Smad7, the main feedback inhibitor of TGF-β signaling in this cell type in mice. METHODS A mouse strain harboring a conditional knockout allele of Smad7 (Smad7fl/fl) with the tamoxifen-inducible inducible Col1a2-CreERT allele was generated and compared with wild-type mice challenged with the cerulein-based model of chronic pancreatitis. RESULTS Pancreatic stellate cells lacking Smad7 had significantly increased collagen I and fibronectin production and showed a higher activation level in vitro. Surprisingly, the fibrotic index in the pancreata of treated conditional knockout mice was only slightly increased, without statistical significance. Except for fibronectin, the expression of different extracellular matrix proteins and the numbers of fibroblasts and inflammatory cells were similar between Smad7-mutant and control mice. CONCLUSIONS There was no clear evidence that the lack of Smad7 in pancreatic stellate cells plays a major role in experimental pancreatitis, at least in the mouse model investigated here.
Collapse
|
12
|
Devocelle A, Lecru L, Ferlicot S, Bessede T, Candelier JJ, Giron-Michel J, François H. IL-15 Prevents Renal Fibrosis by Inhibiting Collagen Synthesis: A New Pathway in Chronic Kidney Disease? Int J Mol Sci 2021; 22:11698. [PMID: 34769128 PMCID: PMC8583733 DOI: 10.3390/ijms222111698] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD), secondary to renal fibrogenesis, is a public health burden. The activation of interstitial myofibroblasts and excessive production of extracellular matrix (ECM) proteins are major events leading to end-stage kidney disease. Recently, interleukin-15 (IL-15) has been implicated in fibrosis protection in several organs, with little evidence in the kidney. Since endogenous IL-15 expression decreased in nephrectomized human allografts evolving toward fibrosis and kidneys in the unilateral ureteral obstruction (UUO) model, we explored IL-15's renoprotective role by pharmologically delivering IL-15 coupled or not with its soluble receptor IL-15Rα. Despite the lack of effects on myofibroblast accumulation, both IL-15 treatments prevented tubulointerstitial fibrosis (TIF) in UUO as characterized by reduced collagen and fibronectin deposition. Moreover, IL-15 treatments inhibited collagen and fibronectin secretion by transforming growth factor-β (TGF-β)-treated primary myofibroblast cultures, demonstrating that the antifibrotic effect of IL-15 in UUO acts, in part, through a direct inhibition of ECM synthesis by myofibroblasts. In addition, IL-15 treatments resulted in decreased expression of monocyte chemoattractant protein 1 (MCP-1) and subsequent macrophage infiltration in UUO. Taken together, our study highlights a major role of IL-15 on myofibroblasts and macrophages, two main effector cells in renal fibrosis, demonstrating that IL-15 may represent a new therapeutic option for CKD.
Collapse
Affiliation(s)
- Aurore Devocelle
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institute of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, 94807 Villejuif, France; (A.D.); (L.L.); (J.-J.C.)
- Orsay-Vallée Campus, Paris-Saclay University, 91190 Gif-sur-Yvette, France
| | - Lola Lecru
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institute of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, 94807 Villejuif, France; (A.D.); (L.L.); (J.-J.C.)
- Orsay-Vallée Campus, Paris-Saclay University, 91190 Gif-sur-Yvette, France
| | - Sophie Ferlicot
- Service d’Anatomopathologie, Hôpital Bicêtre, AP-HP, 94270 Le Kremlin-Bicêtre, France;
| | - Thomas Bessede
- Service d’Urologie, Hôpital Bicêtre, AP-HP, 94270 Le Kremlin-Bicêtre, France;
| | - Jean-Jacques Candelier
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institute of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, 94807 Villejuif, France; (A.D.); (L.L.); (J.-J.C.)
- Orsay-Vallée Campus, Paris-Saclay University, 91190 Gif-sur-Yvette, France
| | - Julien Giron-Michel
- INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institute of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif and CTSA Clamart, 94807 Villejuif, France; (A.D.); (L.L.); (J.-J.C.)
- Orsay-Vallée Campus, Paris-Saclay University, 91190 Gif-sur-Yvette, France
| | - Hélène François
- INSERM UMR_S1155, Tenon Hospital, 75020 Paris, France
- Soins Intensifs Néphrologiques et Rein Aigu (SINRA), Hôpital Tenon, AP-HP, Sorbonne University, 75020 Paris, France
| |
Collapse
|
13
|
Yuan X, Tang WB, Peng L, Chen Y, Tang S, Ge H, Wang X, Xiao X. Elevation of LncRNA ENST00000453774.1 Prevents Renal Fibrosis by Upregulating FBN1, IGF1R, and KLF7. Kidney Blood Press Res 2021; 46:563-573. [PMID: 34614499 DOI: 10.1159/000515624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 03/03/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Transforming growth factor-β (TGF-β), a common outcome of various progressive chronic kidney diseases, can regulate and induce fibrosis. OBJECTIVE The study aimed to identify downstream targets of lncRNA ENST00000453774.1 (lnc453774.1) and outline their functions on the development of renal fibrosis. METHODS HK-2 cells were induced with 5 ng/mL TGF-β1 for 24 h to construct a renal fibrosis cell model. Differentially expressed genes (DEGs) targeted by lnc453774.1 in TGF-β1-induced renal fibrosis were identified using RNA sequencing. The dataset GSE23338 was employed to identify DEGs in 48-h TGF-β1-stimulated human kidney epithelial cells, and these DEGs were intersected with genes in the key module using weighted gene co-expression network analysis to generate key genes associated with renal fibrosis. MicroRNAs (miRs) that had targeting relationship with keys genes and lnc453774.1 were predicted by using Miranda software, and important genes were intersected with key genes that had targeting relationship with these miRs. Key target genes by lnc453774.1 were identified in a protein-protein interaction network among lnc453774.1, important genes, and reported genes related to autophagy, oxidative stress, and cell adhesion. RESULTS Key genes in the key module (turquoise) were intersected with DEGs in the dataset GSE23338 and yielded 20 key genes regulated by lnc453774.1 involved in renal fibrosis. Fourteen miRs had targeting relationship with lnc453774.1 and key genes, and 8 important genes targeted by these 14 miRs were identified. Fibrillin-1 (FBN1), insulin-like growth factor 1 receptor (IGF1R), and Kruppel-like factor 7 (KLF7) were identified to be involved in autophagy, oxidative stress, and cell adhesion and were elevated in the lnc453774.1-overexpressing TGF-β1-induced cells. CONCLUSION These results show FBN1, IGF1R, and KLF7 serve as downstream targets of lnc453774.1, and that lnc453774.1 may protect against renal fibrosis through competing endogenous miRs which target FBN1, IGF1R, and KLF7 mRNAs.
Collapse
Affiliation(s)
- Xiangning Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Bin Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Peng
- The Nephrotic Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Yusa Chen
- Department of Nephrology, Hunan Provincial People's Hospital, Changsha, China
| | - Shumei Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Huipeng Ge
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiufen Wang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Neder TH, Schrankl J, Fuchs MAA, Broeker KAE, Wagner C. Endothelin receptors in renal interstitial cells do not contribute to the development of fibrosis during experimental kidney disease. Pflugers Arch 2021; 473:1667-1683. [PMID: 34355294 PMCID: PMC8433107 DOI: 10.1007/s00424-021-02604-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/24/2021] [Accepted: 07/01/2021] [Indexed: 11/26/2022]
Abstract
Renal interstitial fibrosis is characterized by the development of myofibroblasts, originating from resident renal and immigrating cells. Myofibroblast formation and extracellular matrix production during kidney damage are triggered by various factors. Among these, endothelins have been discussed as potential modulators of renal fibrosis. Utilizing mouse models of adenine nephropathy (AN) and unilateral ureter occlusion (UUO), this study aimed to investigate the contribution of endothelin signaling in stromal mesenchymal resident renal interstitial cells. We found in controls that adenine feeding and UUO caused marked upregulations of endothelin-1 (ET-1) gene expression in endothelial and in tubular cells and a strong upregulation of ETA-receptor (ETA-R) gene expression in interstitial and mesangial cells, while the gene expression of ETB-receptor (ETB-R) did not change. Conditional deletion of ETA-R and ETB-R gene expression in the FoxD1 stromal cell compartment which includes interstitial cells significantly reduced renal ETA-R gene expression and moderately lowered renal ETB-R gene expression. ET receptor (ET-R) deletion exerted no apparent effects on kidney development nor on kidney function. Adenine feeding and UUO led to similar increases in profibrotic and proinflammatory gene expression in control as well as in ETAflflETBflfl FoxD1Cre+ mice (ET-Ko). In summary, our findings suggest that adenine feeding and UUO activate endothelin signaling in interstitial cells which is due to upregulated ETA-R expression and enhanced renal ET-1 production Our data also suggest that the activation of endothelin signaling in interstitial cells has less impact for the development of experimentally induced fibrosis.
Collapse
Affiliation(s)
- Thomas H Neder
- Institute of Physiology, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Julia Schrankl
- Institute of Physiology, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Michaela A A Fuchs
- Institute of Physiology, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Katharina A E Broeker
- Institute of Physiology, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany.
| |
Collapse
|
15
|
Unachukwu U, Shiomi T, Goldklang M, Chada K, D'Armiento J. Renal neoplasms in tuberous sclerosis mice are neurocristopathies. iScience 2021; 24:102684. [PMID: 34222844 PMCID: PMC8243016 DOI: 10.1016/j.isci.2021.102684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/20/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Tuberous sclerosis (TS) is a rare disorder exhibiting multi-systemic benign neoplasms. We hypothesized the origin of TS neoplastic cells derived from the neural crest given the heterogeneous ecto-mesenchymal phenotype of the most common TS neoplasms. To test this hypothesis, we employed Cre-loxP lineage tracing of myelin protein zero (Mpz)-expressing neural crest cells (NCCs) in spontaneously developing renal tumors of Tsc2 +/- /Mpz(Cre)/TdT fl/fl reporter mice. In these mice, ectopic renal tumor onset was detected at 4 months of age increasing in volume by 16 months of age with concomitant increase in the subpopulation of tdTomato+ NCCs from 0% to 6.45% of the total number of renal tumor cells. Our results suggest that Tsc2 +/- mouse renal tumors arise from domiciled proliferative progenitor cell populations of neural crest origin that co-opt tumorigenesis due to mutations in Tsc2 loci. Targeting neural crest antigenic determinants will provide a potential alternative therapeutic approach for TS pathogenesis.
Collapse
Affiliation(s)
- Uchenna Unachukwu
- Center for LAM and Rare Lung Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, 630 West 168 Street, New York, NY 10032, USA
| | - Takayuki Shiomi
- Department of Pathology, International University of Health and Welfare, School of Medicine, 4-3 Kouzunomori, Narita-shi, Chiba 286-8686, Japan
| | - Monica Goldklang
- Center for LAM and Rare Lung Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, 630 West 168 Street, New York, NY 10032, USA
| | - Kiran Chada
- Department of Biochemistry, Rutgers-Robert Wood Johnson Medical School, Rutgers University, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | - Jeanine D'Armiento
- Center for LAM and Rare Lung Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, 630 West 168 Street, New York, NY 10032, USA
| |
Collapse
|
16
|
Luo XM, Yan C, Feng YM. Nanomedicine for the treatment of diabetes-associated cardiovascular diseases and fibrosis. Adv Drug Deliv Rev 2021; 172:234-248. [PMID: 33417981 DOI: 10.1016/j.addr.2021.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/25/2020] [Accepted: 01/01/2021] [Indexed: 02/08/2023]
Abstract
Cardiomyopathy and fibrosis are the main causes of heart failure in diabetes patients. For therapeutic purposes, a delivery system is required to enhance antidiabetic drug efficacy and specifically target profibrotic pathways in cardiomyocytes. Nanoparticles (NPs) have distinct advantages, including biocompatibility, bioavailability, targeting efficiency, and minimal toxicity, which make them ideal for antidiabetic treatment. In this review, we overview the latest information on the pathogenesis of cardiomyopathy and fibrosis in diabetes patients. We summarize how NP applications improve insulin and liraglutide efficacy and their sustained release upon oral administration. We provide a comprehensive review of the results of NP clinical trials in diabetes patients and of animal studies investigating the effects of NP-mediated anti-fibrotic treatments. Collectively, the application of advanced NP delivery systems in the treatment of cardiomyopathy and fibrosis in diabetes patients is a promising and innovative therapeutic strategy.
Collapse
|
17
|
Chen B, Wang P, Liang X, Jiang C, Ge Y, Dworkin LD, Gong R. Permissive effect of GSK3β on profibrogenic plasticity of renal tubular cells in progressive chronic kidney disease. Cell Death Dis 2021; 12:432. [PMID: 33931588 PMCID: PMC8087712 DOI: 10.1038/s41419-021-03709-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
Renal tubular epithelial cells (TECs) play a key role in renal fibrogenesis. After persistent injuries that are beyond self-healing capacity, TECs will dedifferentiate, undergo growth arrest, convert to profibrogenic phenotypes, and resort to maladaptive plasticity that ultimately results in renal fibrosis. Evidence suggests that glycogen synthase kinase (GSK) 3β is centrally implicated in kidney injury. However, its role in renal fibrogenesis is obscure. Analysis of publicly available kidney transcriptome database demonstrated that patients with progressive chronic kidney disease (CKD) exhibited GSK3β overexpression in renal tubulointerstitium, in which the predefined hallmark gene sets implicated in fibrogenesis were remarkably enriched. In vitro, TGF-β1 treatment augmented GSK3β expression in TECs, concomitant with dedifferentiation, cell cycle arrest at G2/M phase, excessive accumulation of extracellular matrix, and overproduction of profibrotic cytokines like PAI-1 and CTGF. All these profibrogenic phenotypes were largely abrogated by GSK3β inhibitors or by ectopic expression of a dominant-negative mutant of GSK3β but reinforced in cells expressing the constitutively active mutant of GSK3β. Mechanistically, GSK3β suppressed, whereas inhibiting GSK3β facilitated, the activity of cAMP response element-binding protein (CREB), which competes for CREB-binding protein, a transcriptional coactivator essential for TGF-β1/Smad signaling pathway to drive TECs profibrogenic plasticity. In vivo, in mice with folic acid-induced progressive CKD, targeting of GSK3β in renal tubules via genetic ablation or by microdose lithium mitigated the profibrogenic plasticity of TEC, concomitant with attenuated interstitial fibrosis and tubular atrophy. Collectively, GSK3β is likely a pragmatic therapeutic target for averting profibrogenic plasticity of TECs and improving renal fibrosis.
Collapse
Affiliation(s)
- Bohan Chen
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, 02903, USA
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Pei Wang
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, 02903, USA
| | - Xianhui Liang
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, 02903, USA
| | - Chunming Jiang
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, 02903, USA
| | - Yan Ge
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, 02903, USA
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Lance D Dworkin
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, 02903, USA
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Rujun Gong
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, 02903, USA.
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, 43614, USA.
| |
Collapse
|
18
|
Lee JH, Park M, Jung K, Hong G, Lee HL, Kim DW, Kim CE, Kang KS. Identification of gallic acid as a active ingredient of Syzygium aromaticum against tacrolimus-induced damage in renal epithelial LLC-PK1 cells and rat kidney. Bioorg Med Chem Lett 2021; 41:128012. [PMID: 33838305 DOI: 10.1016/j.bmcl.2021.128012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 11/29/2022]
Abstract
Tacrolimus (FK506), a calcineurin inhibitor, is an effective immunosuppressive agent mainly used to lower the risk of organ rejection after allogeneic organ transplant. However, FK506-associated adverse effects, such as nephrotoxicity, may limit its therapeutic use. In this study, we confirmed that epigallocatechin-3-gallate (EGCG), sanguiin H-6, and gallic acid increased cell survival following FK506-induced cytotoxicity in renal epithelial LLC-PK1. Among these compounds, gallic acid exerted the strongest protective effect, further confirmed in the FK506-induced nephrotoxicity rat model. Additionally, we identified supporting evidence for the nephroprotective function of gallic acid using molecular docking and bioavailability investigations.
Collapse
Affiliation(s)
- Ji Hwan Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Musun Park
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; Intellectual Information Team, Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Kiwon Jung
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Sungnam 13844, Republic of Korea
| | - Gyeongmin Hong
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Sungnam 13844, Republic of Korea
| | - Hye Lim Lee
- Department of Pediatrics, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| | - Dong-Wook Kim
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28530, Republic of Korea
| | - Chang-Eop Kim
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea.
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea.
| |
Collapse
|
19
|
Fuchs MAA, Broeker KAE, Schrankl J, Burzlaff N, Willam C, Wagner C, Kurtz A. Inhibition of transforming growth factor β1 signaling in resident interstitial cells attenuates profibrotic gene expression and preserves erythropoietin production during experimental kidney fibrosis in mice. Kidney Int 2021; 100:122-137. [PMID: 33705825 DOI: 10.1016/j.kint.2021.02.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 02/10/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022]
Abstract
Kidney fibrosis is characterized by the development of myofibroblasts originating from resident kidney and immigrating cells. Myofibroblast formation and extracellular matrix production during kidney damage are triggered by various cytokines. Among these, transforming growth factor β1 (TGFβ1) is considered a central trigger for kidney fibrosis. We found a highly upregulated expression of TGFβ1 and TGFβ receptor 2 (TGFβ-R2) mRNAs in kidney interstitial cells in experimental fibrosis. Here, we investigated the contribution of TGFβ1 signaling in resident kidney interstitial cells to organ fibrosis using the models of adenine induced nephropathy and unilateral ureteral occlusion in mice. For this purpose TGFβ1 signaling was interrupted by inducible deletion of the TGFβ-R2 gene in interstitial cells expressing the fibroblast marker platelet derived growth factor receptor-β. Expression of profibrotic genes was attenuated up to 50% in kidneys lacking TGFβ-R2 in cells positive for platelet derived growth factor receptor-β. Additionally, deletion of TGFβ-R2 prevented the decline of erythropoietin production in ureter ligated kidneys. Notably, fibrosis associated expression of α-smooth muscle actin as a myofibroblast marker and deposits of extracellular collagens were not altered in mice with targeted deletion of TGFβ-R2. Thus, our findings suggest an enhancing effect of TGFβ1 signaling in resident interstitial cells that contributes to profibrotic gene expression and the downregulation of erythropoietin production, but not to the development of myofibroblasts during kidney fibrosis.
Collapse
Affiliation(s)
| | | | - Julia Schrankl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Nicolai Burzlaff
- Department of Chemistry and Pharmacy, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Carsten Willam
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
20
|
Geng H, Lan R, Liu Y, Chen W, Wu M, Saikumar P, Weinberg JM, Venkatachalam MA. Proximal tubule LPA1 and LPA2 receptors use divergent signaling pathways to additively increase profibrotic cytokine secretion. Am J Physiol Renal Physiol 2021; 320:F359-F374. [PMID: 33427061 PMCID: PMC7988817 DOI: 10.1152/ajprenal.00494.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/17/2020] [Accepted: 12/30/2020] [Indexed: 01/01/2023] Open
Abstract
Lysophosphatidic acid (LPA) increases platelet-derived growth factor-B (PDGFB) and connective tissue growth factor (CTGF) production and secretion by proximal tubule (PT) cells through LPA2 receptor-Gqα-αvβ6-integrin-mediated activation of transforming growth factor-β1 (TGFB1). LPA2, β6-integrin, PDGFB, and CTGF increase in kidneys after ischemia-reperfusion injury (IRI), coinciding with fibrosis. The TGFB1 receptor antagonist SD-208 prevents increases of β6-integrin, TGFB1-SMAD signaling, and PDGFB/CTGF expression after IRI and ameliorates fibrosis (Geng H, Lan R, Singha PK, Gilchrist A, Weinreb PH, Violette SM, Weinberg JM, Saikumar P, Venkatachalam MA. Am J Pathol 181: 1236-1249, 2012; Geng H, Lan R, Wang G, Siddiqi AR, Naski MC, Brooks AI, Barnes JL, Saikumar P, Weinberg JM, Venkatachalam MA. Am J Pathol 174: 1291-1308, 2009). We report now that LPA1 receptor signaling through epidermal growth factor receptor (EGFR)-ERK1/2-activator protein-1 cooperates with LPA2-dependent TGFB1 signaling to additively increase PDGFB/CTGF production and secretion by PT cells. Conversely, inhibition of both pathways results in greater suppression of PDGFB/CTGF production and secretion and promotes greater PT cellular differentiation than inhibiting one pathway alone. Antagonism of the LPA-generating enzyme autotaxin suppressed signaling through both pathways. After IRI, kidneys showed not only more LPA2, nuclear SMAD2/3, and PDGFB/CTGF but also increased LPA1 and autotaxin proteins, together with enhanced EGFR/ERK1/2 activation. Remarkably, the TGFB1 receptor antagonist SD-208 prevented all of these abnormalities excepting increased LPA2. SD-208 inhibits only one arm of LPA signaling: LPA2-Gqα-αvβ6-integrin-dependent production of active TGFB1 and its receptor-bound downstream effects. Consequently, far-reaching protection by SD-208 against IRI-induced signaling alterations and tubule-interstitial pathology is not fully explained by our data. TGFB1-dependent feedforward modulation of LPA1 signaling is one possibility. SD-208 effects may also involve mitigation of injury caused by IRI-induced TGFB1 signaling in endothelial cells and monocytes. Our results have translational implications for using TGFB1 receptor antagonists, LPA1 and LPA2 inhibitors concurrently, and autotaxin inhibitors in acute kidney injury to prevent the development of chronic kidney disease.
Collapse
Affiliation(s)
- Hui Geng
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Rongpei Lan
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Yaguang Liu
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Wei Chen
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Meng Wu
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Pothana Saikumar
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Joel M Weinberg
- Department of Medicine, University of Michigan Medical Center, Ann Arbor, Michigan
| | | |
Collapse
|
21
|
Chen YT, Jhao PY, Hung CT, Wu YF, Lin SJ, Chiang WC, Lin SL, Yang KC. Endoplasmic reticulum protein TXNDC5 promotes renal fibrosis by enforcing TGF-β signaling in kidney fibroblasts. J Clin Invest 2021; 131:143645. [PMID: 33465051 DOI: 10.1172/jci143645] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/13/2021] [Indexed: 01/18/2023] Open
Abstract
Renal fibrosis, a common pathological manifestation of virtually all types of chronic kidney disease (CKD), often results in diffuse kidney scarring and predisposes to end-stage renal disease. Currently, there is no effective therapy against renal fibrosis. Recently, our laboratory identified an ER-resident protein, thioredoxin domain containing 5 (TXNDC5), as a critical mediator of cardiac fibrosis. Transcriptome analyses of renal biopsy specimens from patients with CKD revealed marked TXNDC5 upregulation in fibrotic kidneys, suggesting a potential role of TXNDC5 in renal fibrosis. Employing multiple fluorescence reporter mouse lines, we showed that TXNDC5 was specifically upregulated in collagen-secreting fibroblasts in fibrotic mouse kidneys. In addition, we showed that TXNDC5 was required for TGF-β1-induced fibrogenic responses in human kidney fibroblasts (HKFs), whereas TXNDC5 overexpression was sufficient to promote HKF activation, proliferation, and collagen production. Mechanistically, we showed that TXNDC5, transcriptionally controlled by the ATF6-dependent ER stress pathway, mediated its profibrogenic effects by enforcing TGF-β signaling activity through posttranslational stabilization and upregulation of type I TGF-β receptor in kidney fibroblasts. Using a tamoxifen-inducible, fibroblast-specific Txndc5 knockout mouse line, we demonstrated that deletion of Txndc5 in kidney fibroblasts mitigated the progression of established kidney fibrosis, suggesting the therapeutic potential of TXNDC5 targeting for renal fibrosis and CKD.
Collapse
Affiliation(s)
- Yen-Ting Chen
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Yu Jhao
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chen-Ting Hung
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yueh-Feng Wu
- Research Center for Developmental Biology and Regenerative Medicine and.,Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Sung-Jan Lin
- Research Center for Developmental Biology and Regenerative Medicine and.,Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Wen-Chih Chiang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Research Center for Developmental Biology and Regenerative Medicine and.,Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Integrated Diagnostics and Therapeutics and
| | - Kai-Chien Yang
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine and.,Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
22
|
Chen J, Wang X, He Q, Bulus N, Fogo AB, Zhang MZ, Harris RC. YAP Activation in Renal Proximal Tubule Cells Drives Diabetic Renal Interstitial Fibrogenesis. Diabetes 2020; 69:2446-2457. [PMID: 32843569 PMCID: PMC7576565 DOI: 10.2337/db20-0579] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/14/2020] [Indexed: 12/18/2022]
Abstract
An increasing number of studies suggest that the renal proximal tubule is a site of injury in diabetic nephropathy (DN), and progressive renal tubulointerstitial fibrosis is an important mediator of progressive kidney dysfunction in DN. In this study, we observed increased expression and activation of YAP (yes-associated protein) in renal proximal tubule epithelial cells (RPTC) in patients with diabetes and in mouse kidneys. Inducible deletion of Yap specifically in RPTC or administration of the YAP inhibitor verteporfin significantly attenuated diabetic tubulointerstitial fibrosis. EGFR-dependent activation of RhoA/Rock and PI3K-Akt signals and their reciprocal interaction were upstream of proximal tubule YAP activation in diabetic kidneys. Production and release of CTGF in culture medium were significantly augmented in human embryonic kidney (HEK)-293 cells transfected with a constitutively active YAP mutant, and the conditioned medium collected from these cells activated and transduced fibroblasts into myofibroblasts. This study demonstrates that proximal tubule YAP-dependent paracrine mechanisms play an important role in diabetic interstitial fibrogenesis; therefore, targeting Hippo signaling may be a therapeutic strategy to prevent the development and progression of diabetic interstitial fibrogenesis.
Collapse
Affiliation(s)
- Jianchun Chen
- Department of Veterans Affairs, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Kidney Disease, Nashville, TN
| | - Xiaoyong Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Qian He
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Nada Bulus
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Agnes B Fogo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Ming-Zhi Zhang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Kidney Disease, Nashville, TN
| | - Raymond C Harris
- Department of Veterans Affairs, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Kidney Disease, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
23
|
Ray HC, Corliss BA, Bruce AC, Kesting S, Dey P, Mansour J, Seaman SA, Smolko CM, Mathews C, Dey BK, Owens GK, Peirce SM, Yates PA. Myh11+ microvascular mural cells and derived mesenchymal stem cells promote retinal fibrosis. Sci Rep 2020; 10:15808. [PMID: 32978500 PMCID: PMC7519078 DOI: 10.1038/s41598-020-72875-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 08/10/2020] [Indexed: 12/29/2022] Open
Abstract
Retinal diseases are frequently characterized by the accumulation of excessive scar tissue found throughout the neural retina. However, the pathophysiology of retinal fibrosis remains poorly understood, and the cell types that contribute to the fibrotic response are incompletely defined. Here, we show that myofibroblast differentiation of mural cells contributes directly to retinal fibrosis. Using lineage tracing technology, we demonstrate that after chemical ocular injury, Myh11+ mural cells detach from the retinal microvasculature and differentiate into myofibroblasts to form an epiretinal membrane. Inhibition of TGFβR attenuates Myh11+ retinal mural cell myofibroblast differentiation, and diminishes the subsequent formation of scar tissue on the surface of the retina. We demonstrate retinal fibrosis within a murine model of oxygen-induced retinopathy resulting from the intravitreal injection of adipose Myh11-derived mesenchymal stem cells, with ensuing myofibroblast differentiation. In this model, inhibiting TGFβR signaling does not significantly alter myofibroblast differentiation and collagen secretion within the retina. This work shows the complexity of retinal fibrosis, where scar formation is regulated both by TGFβR and non-TGFβR dependent processes involving mural cells and derived mesenchymal stem cells. It also offers a cautionary note on the potential deleterious, pro-fibrotic effects of exogenous MSCs once intravitreally injected into clinical patients.
Collapse
Affiliation(s)
- H Clifton Ray
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Bruce A Corliss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Anthony C Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Sam Kesting
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Paromita Dey
- The RNA Institute, University at Albany, State University of New York, Albany, NY, USA
| | - Jennifer Mansour
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Scott A Seaman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Christian M Smolko
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Corbin Mathews
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Bijan K Dey
- The RNA Institute, University at Albany, State University of New York, Albany, NY, USA
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Paul A Yates
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
- Department of Ophthalmology, University of Virginia, PO Box 800715, Charlottesville, VA, 22908, USA.
| |
Collapse
|
24
|
Yan Q, Zhu K, Zhang L, Fu Q, Chen Z, Liu S, Fu D, Nakazato R, Yoshioka K, Diao B, Ding G, Li X, Wang H. A negative feedback loop between JNK-associated leucine zipper protein and TGF-β1 regulates kidney fibrosis. Commun Biol 2020; 3:288. [PMID: 32504044 PMCID: PMC7275040 DOI: 10.1038/s42003-020-1008-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 05/17/2020] [Indexed: 12/26/2022] Open
Abstract
Renal fibrosis is controlled by profibrotic and antifibrotic forces. Exploring anti-fibrosis factors and mechanisms is an attractive strategy to prevent organ failure. Here we identified the JNK-associated leucine zipper protein (JLP) as a potential endogenous antifibrotic factor. JLP, predominantly expressed in renal tubular epithelial cells (TECs) in normal human or mouse kidneys, was downregulated in fibrotic kidneys. Jlp deficiency resulted in more severe renal fibrosis in unilateral ureteral obstruction (UUO) mice, while renal fibrosis resistance was observed in TECs-specific transgenic Jlp mice. JLP executes its protective role in renal fibrosis via negatively regulating TGF-β1 expression and autophagy, and the profibrotic effects of ECM production, epithelial-to-mesenchymal transition (EMT), apoptosis and cell cycle arrest in TECs. We further found that TGF-β1 and FGF-2 could negatively regulate the expression of JLP. Our study suggests that JLP plays a central role in renal fibrosis via its negative crosstalk with the profibrotic factor, TGF-β1. Qi Yan et al. find that JNK-associated leucine zipper protein (Jlp) counteracts the profibrotic effects of TGF-β1 and autophagy on renal tubular epithelial cells and that TGF-β1 and FGF-2 can negatively regulate the expression of Jlp. These findings provide insights into the role of Jlp in kidney fibrosis.
Collapse
Affiliation(s)
- Qi Yan
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhu
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Internal Medicine, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Qiang Fu
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaowei Chen
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shan Liu
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dou Fu
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ryota Nakazato
- Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Katsuji Yoshioka
- Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Bo Diao
- Department of Medical Laboratory Center, General Hospital of Central Theater Command, Wuhan, China
| | - Guohua Ding
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaogang Li
- Department of Internal Medicine, and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Huiming Wang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
25
|
Hu C, Lakshmipathi J, Stuart D, Peti-Peterdi J, Gyarmati G, Hao CM, Hansell P, Kohan DE. Renomedullary Interstitial Cell Endothelin A Receptors Regulate BP and Renal Function. J Am Soc Nephrol 2020; 31:1555-1568. [PMID: 32487560 DOI: 10.1681/asn.2020020232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/06/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The physiologic role of renomedullary interstitial cells, which are uniquely and abundantly found in the renal inner medulla, is largely unknown. Endothelin A receptors regulate multiple aspects of renomedullary interstitial cell function in vitro. METHODS To assess the effect of targeting renomedullary interstitial cell endothelin A receptors in vivo, we generated a mouse knockout model with inducible disruption of renomedullary interstitial cell endothelin A receptors at 3 months of age. RESULTS BP and renal function were similar between endothelin A receptor knockout and control mice during normal and reduced sodium or water intake. In contrast, on a high-salt diet, compared with control mice, the knockout mice had reduced BP; increased urinary sodium, potassium, water, and endothelin-1 excretion; increased urinary nitrite/nitrate excretion associated with increased noncollecting duct nitric oxide synthase-1 expression; increased PGE2 excretion associated with increased collecting duct cyclooxygenase-1 expression; and reduced inner medullary epithelial sodium channel expression. Water-loaded endothelin A receptor knockout mice, compared with control mice, had markedly enhanced urine volume and reduced urine osmolality associated with increased urinary endothelin-1 and PGE2 excretion, increased cyclooxygenase-2 protein expression, and decreased inner medullary aquaporin-2 protein content. No evidence of endothelin-1-induced renomedullary interstitial cell contraction was observed. CONCLUSIONS Disruption of renomedullary interstitial cell endothelin A receptors reduces BP and increases salt and water excretion associated with enhanced production of intrinsic renal natriuretic and diuretic factors. These studies indicate that renomedullary interstitial cells can modulate BP and renal function under physiologic conditions.
Collapse
Affiliation(s)
- Chunyan Hu
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| | | | - Deborah Stuart
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| | - Janos Peti-Peterdi
- Departments of Physiology and Neuroscience and Medicine, University of Southern California, Los Angeles, California
| | - Georgina Gyarmati
- Departments of Physiology and Neuroscience and Medicine, University of Southern California, Los Angeles, California
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter Hansell
- Department of Medical Cell Biology, Section of Integrative Physiology, Uppsala University Biomedical Center, Uppsala, Sweden
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| |
Collapse
|
26
|
Nlandu-Khodo S, Osaki Y, Scarfe L, Yang H, Phillips-Mignemi M, Tonello J, Saito-Diaz K, Neelisetty S, Ivanova A, Huffstater T, McMahon R, Taketo MM, deCaestecker M, Kasinath B, Harris RC, Lee E, Gewin LS. Tubular β-catenin and FoxO3 interactions protect in chronic kidney disease. JCI Insight 2020; 5:135454. [PMID: 32369448 DOI: 10.1172/jci.insight.135454] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays an important role in renal development and is reexpressed in the injured kidney and other organs. β-Catenin signaling is protective in acute kidney injury (AKI) through actions on the proximal tubule, but the current dogma is that Wnt/β-catenin signaling promotes fibrosis and development of chronic kidney disease (CKD). As the role of proximal tubular β-catenin signaling in CKD remains unclear, we genetically stabilized (i.e., activated) β-catenin specifically in murine proximal tubules. Mice with increased tubular β-catenin signaling were protected in 2 murine models of AKI to CKD progression. Oxidative stress, a common feature of CKD, reduced the conventional T cell factor/lymphoid enhancer factor-dependent β-catenin signaling and augmented FoxO3-dependent activity in proximal tubule cells in vitro and in vivo. The protective effect of proximal tubular β-catenin in renal injury required the presence of FoxO3 in vivo. Furthermore, we identified cystathionine γ-lyase as a potentially novel transcriptional target of β-catenin/FoxO3 interactions in the proximal tubule. Thus, our studies overturned the conventional dogma about β-catenin signaling and CKD by showing a protective effect of proximal tubule β-catenin in CKD and identified a potentially new transcriptional target of β-catenin/FoxO3 signaling that has therapeutic potential for CKD.
Collapse
Affiliation(s)
- Stellor Nlandu-Khodo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA.,Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Yosuke Osaki
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Lauren Scarfe
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, Tennessee, USA
| | - Melanie Phillips-Mignemi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Jane Tonello
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | | | - Surekha Neelisetty
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Alla Ivanova
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Tessa Huffstater
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Robert McMahon
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - M Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mark deCaestecker
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | - Balakuntalam Kasinath
- Department of Medicine, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA.,Department of Medicine, Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Ethan Lee
- Department of Cell and Developmental Biology and
| | - Leslie S Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA.,Department of Cell and Developmental Biology and.,Department of Medicine, Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
27
|
Yao L, Wright MF, Farmer BC, Peterson LS, Khan AM, Zhong J, Gewin L, Hao CM, Yang HC, Fogo AB. Fibroblast-specific plasminogen activator inhibitor-1 depletion ameliorates renal interstitial fibrosis after unilateral ureteral obstruction. Nephrol Dial Transplant 2020; 34:2042-2050. [PMID: 31071225 DOI: 10.1093/ndt/gfz050] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 02/20/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1) expression increases extracellular matrix deposition and contributes to interstitial fibrosis in the kidney after injury. While PAI-1 is ubiquitously expressed in the kidney, we hypothesized that interstitial fibrosis is strongly dependent on fibroblast-specific PAI-1 (fbPAI-1). METHODS Tenascin C Cre (TNC Cre) and fbPAI-1 knockdown (KD) mice with green fluorescent protein (GFP) expressed within the TNC construct underwent unilateral ureteral obstruction and were sacrificed 10 days later. RESULTS GFP+ cells in fbPAI-1 KD mice showed significantly reduced PAI-1 expression. Interstitial fibrosis, measured by Sirius red staining and collagen I western blot, was significantly decreased in fbPAI-1 KD compared with TNC Cre mice. There was no significant difference in transforming growth factor β (TGF-β) expression or its activation between the two groups. However, GFP+ cells from fbPAI-1 KD mice had lower TGF β and connective tissue growth factor (CTGF) expression. The number of fibroblasts was decreased in fbPAI-1 KD compared with TNC Cre mice, correlating with decreased alpha smooth muscle actin (α-SMA) expression and less fibroblast cell proliferation. TNC Cre mice had decreased E-cadherin, a marker of differentiated tubular epithelium, in contrast to preserved expression in fbPAI-1 KD. F4/80-expressing cells, mostly CD11c+/F4/80+ cells, were increased while M1 macrophage markers were decreased in fbPAI-1 KD compared with TNC Cre mice. CONCLUSION These findings indicate that fbPAI-1 depletion ameliorates interstitial fibrosis by decreasing fibroblast proliferation in the renal interstitium, with resulting decreased collagen I. This is linked to decreased M1 macrophages and preserved tubular epithelium.
Collapse
Affiliation(s)
- Lan Yao
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Medical Healthcare Center, Beijing Friendship Hospital of Capital Medical University, Beijing, China
| | - M Frances Wright
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brandon C Farmer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biology, Western Kentucky University, Bowling Green, KY, USA
| | - Laura S Peterson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amir M Khan
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Jianyong Zhong
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leslie Gewin
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hai-Chun Yang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Agnes B Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA.,Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
28
|
Li H, Shao F, Qian B, Sun Y, Huang Z, Ding Z, Dong L, Chen J, Zhang J, Zang Y. Upregulation of HER2 in tubular epithelial cell drives fibroblast activation and renal fibrosis. Kidney Int 2019; 96:674-688. [DOI: 10.1016/j.kint.2019.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 03/12/2019] [Accepted: 04/05/2019] [Indexed: 12/20/2022]
|
29
|
Li X, Clappier C, Kleiter I, Heuchel R. Tamoxifen affects chronic pancreatitis-related fibrogenesis in an experimental mouse model: an effect beyond Cre recombination. FEBS Open Bio 2019; 9:1756-1768. [PMID: 31380604 PMCID: PMC6768287 DOI: 10.1002/2211-5463.12714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/13/2019] [Accepted: 08/02/2019] [Indexed: 01/20/2023] Open
Abstract
Tamoxifen is very successfully used for the induction of CreERT‐mediated genomic recombination in conditional mouse models. Recent studies, however, indicated that tamoxifen might also affect the fibrotic response in several disease models following administration, both in vitro and in vivo. In order to investigate a possible effect of tamoxifen on pancreatic fibrogenesis and to evaluate an optimal treatment scheme in an experimental pancreatitis mouse model, we administered tamoxifen by oral gavage to both male and female C57BL/6J mice and then waited for different periods of time before inducing chronic pancreatitis by cerulein. We observed a sex‐specific and time‐dependent effect of tamoxifen on the fibrotic response as measured by collagen deposition and the number of myofibroblasts and macrophages. The findings of in vitro studies, in which cerulein was administrated with or without 4‐hydroxytamoxifen to stimulate primary murine female and male pancreatic stellate cells, supported our in vivo observations. Real‐time PCR also indicated that this effect may be related to differences in ERα expression between female and male stellate cells. Our data demonstrate that tamoxifen administration has unignorable side effects, which affect the experimental outcome in a cerulein‐based model of chronic pancreatitis in mice. We suggest a 2‐week waiting period before cerulein administration to reduce side effects to a minimum for the described fibrosis model in female mice.
Collapse
Affiliation(s)
- Xuan Li
- Pancreas Cancer Research (PaCaRes) Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Clappier
- Pancreas Cancer Research (PaCaRes) Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ingo Kleiter
- Department of Neurology, Ruhr-Universität Bochum, Germany
| | - Rainer Heuchel
- Pancreas Cancer Research (PaCaRes) Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Zuo Z, Huang P, Jiang Y, Zhang Y, Zhu M. Acupuncture attenuates renal interstitial fibrosis via the TGF‑β/Smad pathway. Mol Med Rep 2019; 20:2267-2275. [PMID: 31322212 PMCID: PMC6691233 DOI: 10.3892/mmr.2019.10470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 03/20/2019] [Indexed: 01/20/2023] Open
Abstract
Acupuncture is one of the most useful tools in complimentary medicine, and has demonstrated potential value for treating chronic renal failure (CRF). However, the underlying mechanisms for its therapeutic effect remain unknown. In the present study, the effects of acupuncture on renal interstitial fibrosis (RIF) were explored in a rabbit model of CRF. Rabbits were assigned to the following five groups: sham, model, losartan potassium (Posi), acupuncture (Acup) and acupuncture+inhibitor (Acup+Inhib) groups. The CRF rabbits were administered a drug or/and acupuncture on Shenshu, Mingmen and Pishu. The body weights, urine protein, serum creatinine (SCr) and blood urea nitrogen (BUN) levels of the rabbits were measured. Transforming growth factor β (TGF-β), integrin-linked kinase (ILK) and Smad3 expression were detected by qRT-PCR. Tumor necrosis factor-α (TNF-α) and endothelial nitric oxide synthase (eNOS) expression were analyzed by western blot methods. The concentrations of TGF-β, IL-8, TNF-α and IL-1β in blood serum were detected using ELISA kits. In addition, pathological characteristics of the rabbit tissues were evaluated by H&E and Masson's trichrome staining methods, and TGF-β expression was detected by immunohistochemistry (IHC) assays. Results showing decreased body weights and increased urine protein, SCr and BUN levels confirmed that the CRF model had been successfully constructed. It was also found that acupuncture significantly reduced the levels of TNF-α, Smad3, ILK and TGF-β expression, dramatically decreased the concentrations of TGF-β, IL-8, TNF-α and IL-1β in blood serum, and significantly increased eNOS expression in the CRF model rabbits by affecting the TGF-β/Smad signaling pathway. In addition, it was demonstrated that acupuncture could relieve RIF by affecting the TGF-β/Smad pathway. These observations indicate that acupuncture may be useful for treating CRF, and suggest the TGF-β/Smad pathway as a target for CRF therapy.
Collapse
Affiliation(s)
- Zheng Zuo
- Acupuncture and Massage College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Peidong Huang
- Acupuncture and Massage Rehabilitation College, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yunwu Jiang
- Acupuncture and Massage Rehabilitation College, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yi Zhang
- Acupuncture and Moxibustion Department, Zibo Hospital of Traditional Chinese Medicine of Shandong Province, Zibo, Shandong 255300, P.R. China
| | - Miansheng Zhu
- Acupuncture and Massage College, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| |
Collapse
|
31
|
Zhang MZ, Wang S, Wang Y, Zhang Y, Ming Hao C, Harris RC. Renal Medullary Interstitial COX-2 (Cyclooxygenase-2) Is Essential in Preventing Salt-Sensitive Hypertension and Maintaining Renal Inner Medulla/Papilla Structural Integrity. Hypertension 2019; 72:1172-1179. [PMID: 30354807 DOI: 10.1161/hypertensionaha.118.11694] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
COX (cyclooxygenase)-derived prostaglandins regulate renal hemodynamics and salt and water homeostasis. Inhibition of COX activity causes blood pressure elevation. In addition, chronic analgesic abuse can induce renal injury, including papillary necrosis. COX-2 is highly expressed in the kidney papilla in renal medullary interstitial cells (RMICs). However, its role in blood pressure and papillary integrity in vivo has not been definitively studied. In mice with selective, inducible RMIC COX-2 deletion, a high-salt diet led to an increase in blood pressure that peaked at 4 to 5 weeks and was associated with increased papillary expression of AQP2 (aquaporin 2) and ENac (epithelial sodium channel) and decreased expression of cystic fibrosis transmembrane conductance regulator. With continued high-salt feeding, the mice with RMIC COX-2 deletion had progressive decreases in blood pressure from its peak. After return to a normal-salt diet for 3 weeks, blood pressure remained low and was associated with a persistent urinary concentrating defect. Within 2 weeks of institution of a high-salt diet, increased apoptotic RMICs and collecting duct cells could be detected in papillae with RMIC deletion of COX-2, and by 9 weeks of high salt, there was a striking loss of the papillae. Therefore, RMIC COX-2 expression plays a crucial role in renal handling water and sodium homeostasis, preventing salt-sensitive hypertension and maintaining structural integrity of papilla.
Collapse
Affiliation(s)
- Ming-Zhi Zhang
- From the Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., Y.Z., R.C.H.).,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., R.C.H.)
| | - Suwan Wang
- From the Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., Y.Z., R.C.H.).,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., R.C.H.)
| | - Yinqiu Wang
- From the Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., Y.Z., R.C.H.).,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., R.C.H.)
| | - Yahua Zhang
- From the Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., Y.Z., R.C.H.)
| | - Chuan Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China (C.M.H.)
| | - Raymond C Harris
- From the Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., Y.Z., R.C.H.).,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN (M.-Z.Z., S.W., Y.W., R.C.H.).,Department of Veterans Affairs, Nashville, TN (R.C.H.)
| |
Collapse
|
32
|
Manolopoulou M, Matlock BK, Nlandu-Khodo S, Simmons AJ, Lau KS, Phillips-Mignemi M, Ivanova A, Alford CE, Flaherty DK, Gewin LS. Novel kidney dissociation protocol and image-based flow cytometry facilitate improved analysis of injured proximal tubules. Am J Physiol Renal Physiol 2019; 316:F847-F855. [PMID: 30759021 PMCID: PMC6580245 DOI: 10.1152/ajprenal.00354.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 12/16/2022] Open
Abstract
Flow cytometry studies on injured kidney tubules are complicated by the low yield of nucleated single cells. Furthermore, cell-specific responses such as cell cycle dynamics in vivo have conventionally relied on indirect immunohistochemistry and proximal tubule markers that may be downregulated in injury. Here, we report a new tissue dissociation protocol for the kidney with an early fixation step that greatly enhances the yield of single cells. Genetic labeling of the proximal tubule with either mT/mG "tomato" or R26Fucci2aR (Fucci) cell cycle reporter mice allows us to follow proximal tubule-specific changes in cell cycle after renal injury. Image-based flow cytometry (FlowSight) enables gating of the cell cycle and concurrent visualization of the cells with bright field and fluorescence. We used the Fucci mouse in conjunction with FlowSight to identify a discrete polyploid population in proximal tubules after aristolochic acid injury. The tissue dissociation protocol in conjunction with genetic labeling and image-based flow cytometry is a tool that can improve our understanding of any discrete cell population after injury.
Collapse
Affiliation(s)
- Marika Manolopoulou
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Brittany K Matlock
- Flow Cytometry Shared Resource, Vanderbilt Vaccine Center, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Stellor Nlandu-Khodo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Alan J Simmons
- Epithelial Biology Center, Vanderbilt University Medical Center , Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
| | - Ken S Lau
- Epithelial Biology Center, Vanderbilt University Medical Center , Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center , Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
- Program in Chemical and Physical Biology, Vanderbilt University , Nashville, Tennessee
| | - Melanie Phillips-Mignemi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Alla Ivanova
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Catherine E Alford
- Department of Pathology and Laboratory Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - David K Flaherty
- Flow Cytometry Shared Resource, Vanderbilt Vaccine Center, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Leslie S Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
- Department of Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
| |
Collapse
|
33
|
El-Deeb OS, Atef MM, Hafez YM. The interplay between microbiota-dependent metabolite trimethylamine N-oxide, Transforming growth factor β/SMAD signaling and inflammasome activation in chronic kidney disease patients: A new mechanistic perspective. J Cell Biochem 2019; 120:14476-14485. [PMID: 31002427 DOI: 10.1002/jcb.28707] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/15/2019] [Accepted: 01/24/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) signifies a frequently life-threatening condition influencing kidney structure and function. Despite its irrefutable importance, its exact pathogenesis is not completely clarified. However, CKD is known to be associated with accumulated uremic toxins/metabolites, interstitial fibrosis, and systemic inflammation. So we aimed to investigate the role of microbiota-dependent metabolite trimethylamine N-oxide (TMAO), transforming growth factor β (TGFβ)/SMAD signaling, and inflammasome activation in CKD pathogenesis through its different stages. SUBJECTS AND METHODS Eighty patients with CKD of stages 2 to 4 in addition 15 healthy control subjects were enrolled. SMAD3 and nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) messenger RNA (mRNA) expressions from whole blood were assessed by quantitative real-time polymerase chain reaction (RT-PCR). Serum TGF-β1 and interleukin-1β (IL-1β) levels were estimated by the enzyme-linked immunosorbent assay. Plasma and urinary TMAO levels were measured. Oxidative stress markers were also assessed. RESULTS SMAD3 and NLRP3 mRNA expressions were significantly upregulated in patients with CKD. Likewise, serum TGF-β1 and IL-1β levels were significantly elevated in patients with CKD, with increase in plasma and urinary TMAO levels and altered redox status throughout different CKD stages. CONCLUSION The study documented that TMAO could be used as a reliable biomarker to evaluate CKD progression; being linked to TGF-β/SMAD signaling, NLRP3 inflammasome activation as well as being a noninvasive applicable technique.
Collapse
Affiliation(s)
- Omnia Safwat El-Deeb
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Marwa Mohamed Atef
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Yasser Mostafa Hafez
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
34
|
Transforming growth factor β (TGFβ) and related molecules in chronic kidney disease (CKD). Clin Sci (Lond) 2019; 133:287-313. [DOI: 10.1042/cs20180438] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/04/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
|
35
|
Chung S, Overstreet JM, Li Y, Wang Y, Niu A, Wang S, Fan X, Sasaki K, Jin GN, Khodo SN, Gewin L, Zhang MZ, Harris RC. TGF-β promotes fibrosis after severe acute kidney injury by enhancing renal macrophage infiltration. JCI Insight 2018; 3:123563. [PMID: 30385721 DOI: 10.1172/jci.insight.123563] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/19/2018] [Indexed: 12/28/2022] Open
Abstract
TGF-β signals through a receptor complex composed of 2 type I and 2 type II (TGF-βRII) subunits. We investigated the role of macrophage TGF-β signaling in fibrosis after AKI in mice with selective monocyte/macrophage TGF-βRII deletion (macrophage TGF-βRII-/- mice). Four weeks after injury, renal TGF-β1 expression and fibrosis were higher in WT mice than macrophage TGF-βRII-/- mice, which had decreased renal macrophages. The in vitro chemotactic response to f-Met-Leu-Phe was comparable between bone marrow-derived monocytes (BMMs) from WT and macrophage TGF-βRII-/- mice, but TGF-βRII-/- BMMs did not respond to TGF-β. We then implanted Matrigel plugs suffused with either f-Met-Leu-Phe or TGF-β1 into WT or macrophage TGF-βRII-/- mice. After 6 days, f-Met-Leu-Phe induced similar macrophage infiltration into the Matrigel plugs of WT and macrophage TGF-βRII-/- mice, but TGF-β induced infiltration only in WT mice. We further determined the number of labeled WT or TGF-βRII-/- BMMs infiltrating into WT kidneys 20 days after ischemic injury. There were more labeled WT BMMs than TGF-βRII-/- BMMs. Therefore, macrophage TGF-βRII deletion protects against the development of tubulointerstitial fibrosis following severe ischemic renal injury. Chemoattraction of macrophages to the injured kidney through a TGF-β/TGF-βRII axis is a heretofore undescribed mechanism by which TGF-β can mediate renal fibrosis during progressive renal injury.
Collapse
Affiliation(s)
- Sungjin Chung
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jessica M Overstreet
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Yan Li
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Yinqiu Wang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Aolei Niu
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Suwan Wang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Xiaofeng Fan
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kensuke Sasaki
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Guan-Nan Jin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Stellor Nlandu Khodo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Leslie Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Veterans Affairs, Nashville, Tennessee, USA
| |
Collapse
|
36
|
TSC1 deletion in fibroblasts alleviates lipopolysaccharide-induced acute kidney injury. Clin Sci (Lond) 2018; 132:2087-2101. [PMID: 30185506 DOI: 10.1042/cs20180348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 11/17/2022]
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) signaling is active in inflammation, but its involvement in septic acute kidney injury (AKI) has not been shown. mTORC1 activation (p-S6) in renal fibroblasts was increased in a mouse AKI model induced by 1.5 mg/kg lipopolysaccharide (LPS). Deletion of tuberous sclerosis complex 1 (TSC1), an mTORC1 negative regulator, in fibroblasts (Fibro-TSC1-/-) inhibited the elevation of serum creatinine and blood urea nitrogen in AKI compared with that in TSC1fl/fl control mice. Endothelin-1 (EDN1) and phospho-Jun-amino-terminal kinase (p-JNK) were up-regulated in Fibro-TSC1-/- renal fibroblasts after LPS challenge. Rapamycin, an mTORC1 inhibitor, and bosentan, an EDN1 antagonist, eliminated the difference in renal function between TSC1fl/fl and Fibro-TSC1-/- mice after LPS injection. Rapamycin restored LPS-induced up-regulation of EDN1, endothelin converting enzyme-1 (ECE1), and p-JNK in TSC1-knockdown mouse embryonic fibroblasts (MEFs). SP600125, a Jun-amino-terminal kinase (JNK) inhibitor, attenuated LPS-induced enhancement of EDN1 and ECE1 in TSC1-knockdown MEFs without a change in phospho-S6 ribosomal protein (p-S6) level. The results indicate that mTORC1-JNK-dependent up-regulation of ECE1 elevated EDN1 in TSC1-knockout renal fibroblasts and contributed to improvement of renal function in Fibro-TSC1-/- mice with LPS-induced AKI. Renal fibroblast mTORC1 plays an important role in septic AKI.
Collapse
|
37
|
Chan SC, Zhang Y, Shao A, Avdulov S, Herrera J, Aboudehen K, Pontoglio M, Igarashi P. Mechanism of Fibrosis in HNF1B-Related Autosomal Dominant Tubulointerstitial Kidney Disease. J Am Soc Nephrol 2018; 29:2493-2509. [PMID: 30097458 DOI: 10.1681/asn.2018040437] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/12/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Mutation of HNF1B, the gene encoding transcription factor HNF-1β, is one cause of autosomal dominant tubulointerstitial kidney disease, a syndrome characterized by tubular cysts, renal fibrosis, and progressive decline in renal function. HNF-1β has also been implicated in epithelial-mesenchymal transition (EMT) pathways, and sustained EMT is associated with tissue fibrosis. The mechanism whereby mutated HNF1B leads to tubulointerstitial fibrosis is not known. METHODS To explore the mechanism of fibrosis, we created HNF-1β-deficient mIMCD3 renal epithelial cells, used RNA-sequencing analysis to reveal differentially expressed genes in wild-type and HNF-1β-deficient mIMCD3 cells, and performed cell lineage analysis in HNF-1β mutant mice. RESULTS The HNF-1β-deficient cells exhibited properties characteristic of mesenchymal cells such as fibroblasts, including spindle-shaped morphology, loss of contact inhibition, and increased cell migration. These cells also showed upregulation of fibrosis and EMT pathways, including upregulation of Twist2, Snail1, Snail2, and Zeb2, which are key EMT transcription factors. Mechanistically, HNF-1β directly represses Twist2, and ablation of Twist2 partially rescued the fibroblastic phenotype of HNF-1β mutant cells. Kidneys from HNF-1β mutant mice showed increased expression of Twist2 and its downstream target Snai2. Cell lineage analysis indicated that HNF-1β mutant epithelial cells do not transdifferentiate into kidney myofibroblasts. Rather, HNF-1β mutant epithelial cells secrete high levels of TGF-β ligands that activate downstream Smad transcription factors in renal interstitial cells. CONCLUSIONS Ablation of HNF-1β in renal epithelial cells leads to the activation of a Twist2-dependent transcriptional network that induces EMT and aberrant TGF-β signaling, resulting in renal fibrosis through a cell-nonautonomous mechanism.
Collapse
Affiliation(s)
| | - Ying Zhang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota; and
| | | | | | | | | | - Marco Pontoglio
- Department of Development, Reproduction and Cancer, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016/Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris-Descartes, Paris, France
| | | |
Collapse
|
38
|
Gewin LS. Renal fibrosis: Primacy of the proximal tubule. Matrix Biol 2018; 68-69:248-262. [PMID: 29425694 PMCID: PMC6015527 DOI: 10.1016/j.matbio.2018.02.006] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 12/20/2022]
Abstract
Tubulointerstitial fibrosis (TIF) is the hallmark of chronic kidney disease and best predictor of renal survival. Many different cell types contribute to TIF progression including tubular epithelial cells, myofibroblasts, endothelia, and inflammatory cells. Previously, most of the attention has centered on myofibroblasts given their central importance in extracellular matrix production. However, emerging data focuses on how the response of the proximal tubule, a specialized epithelial segment vulnerable to injury, plays a central role in TIF progression. Several proximal tubular responses such as de-differentiation, cell cycle changes, autophagy, and metabolic changes may be adaptive initially, but can lead to maladaptive responses that promote TIF both through autocrine and paracrine effects. This review discusses the current paradigm of TIF progression and the increasingly important role of the proximal tubule in promoting TIF both in tubulointerstitial and glomerular injuries. A better understanding and appreciation of the role of the proximal tubule in TIF has important implications for therapeutic strategies to halt chronic kidney disease progression.
Collapse
Affiliation(s)
- Leslie S Gewin
- The Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, United States.
| |
Collapse
|
39
|
Zeng F, Miyazawa T, Kloepfer LA, Harris RC. ErbB4 deletion accelerates renal fibrosis following renal injury. Am J Physiol Renal Physiol 2018; 314:F773-F787. [PMID: 28724608 PMCID: PMC6031915 DOI: 10.1152/ajprenal.00260.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 12/14/2022] Open
Abstract
Tubulointerstitial fibrosis (TIF) is a prominent factor in the progression of chronic kidney disease regardless of etiology. Avian erythroblastic leukemia viral oncogene homolog 4 (ErbB4) expression levels were inversely correlated to renal fibrosis in human fibrotic kidneys. In both unilateral ureteral obstruction (UUO) and ischemia-reperfusion injury followed by uninephrectomy (IRI/UNx) mouse models, expression levels of ErbB4 were elevated in the early stage of renal injury. Using mice with global ErbB4 deletion except for transgenic rescue in cardiac tissue ( ErbB4-/-ht+), we determined that UUO induced similar injury in proximal tubules compared with wild-type mice but more severe injury in distal nephrons. TIF was apparent earlier and was more pronounced following UUO in ErbB4-/-ht+ mice. With ErbB4 deletion, UUO injury inhibited protein kinase B phosphorylation and increased the percentage of cells in G2/M arrest. There was also increased nuclear immunostaining of yes-associated protein and increased expression of phospho-Mothers against decapentaplegic homolog 3, snail1, and vimentin. These results indicate that ErbB4 deletion accelerates the development and progression of renal fibrosis in obstructive nephropathy. Similar results were found in a mouse IRI/UNx model. In conclusion, increased expression of ErbB4 in the early stages of renal injury may reflect a compensatory effect to lessen tubulointerstitial injury.
Collapse
MESH Headings
- Acute Kidney Injury/etiology
- Acute Kidney Injury/genetics
- Acute Kidney Injury/metabolism
- Acute Kidney Injury/pathology
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Case-Control Studies
- Cell Cycle Proteins
- Cell Dedifferentiation
- Disease Models, Animal
- Disease Progression
- Fibrosis
- G2 Phase Cell Cycle Checkpoints
- Gene Deletion
- Genetic Predisposition to Disease
- Kidney/metabolism
- Kidney/pathology
- Mice, Knockout
- Nephrectomy
- Phenotype
- Phosphoproteins/metabolism
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, ErbB-4/deficiency
- Receptor, ErbB-4/genetics
- Receptor, ErbB-4/metabolism
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Reperfusion Injury/etiology
- Reperfusion Injury/genetics
- Reperfusion Injury/metabolism
- Reperfusion Injury/pathology
- Severity of Illness Index
- Signal Transduction
- Smad3 Protein/metabolism
- Snail Family Transcription Factors/metabolism
- Time Factors
- Ureteral Obstruction/complications
- Vimentin/metabolism
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Fenghua Zeng
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Tomoki Miyazawa
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Lance A Kloepfer
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
- Department of Veterans Affairs , Nashville, Tennessee
| |
Collapse
|
40
|
Microvascular Mural Cell Organotypic Heterogeneity and Functional Plasticity. Trends Cell Biol 2018; 28:302-316. [DOI: 10.1016/j.tcb.2017.12.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 01/28/2023]
|
41
|
Lawson JS, Syme HM, Wheeler-Jones CPD, Elliott J. Characterisation of feline renal cortical fibroblast cultures and their transcriptional response to transforming growth factor β1. BMC Vet Res 2018. [PMID: 29523136 PMCID: PMC5845201 DOI: 10.1186/s12917-018-1387-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background Chronic kidney disease (CKD) is common in geriatric cats, and the most prevalent pathology is chronic tubulointerstitial inflammation and fibrosis. The cell type predominantly responsible for the production of extra-cellular matrix in renal fibrosis is the myofibroblast, and fibroblast to myofibroblast differentiation is probably a crucial event. The cytokine TGF-β1 is reportedly the most important regulator of myofibroblastic differentiation in other species. The aim of this study was to isolate and characterise renal fibroblasts from cadaverous kidney tissue of cats with and without CKD, and to investigate the transcriptional response to TGF-β1. Results Cortical fibroblast cultures were successfully established from the kidney tissue of cats with normal kidney function (FCF) and cats with chronic kidney disease (CKD-FCF). Both cell types expressed the mesenchymal markers vimentin, CD44 and CD29, and were negative for the epithelial marker cytokeratin, mesangial cell marker desmin and endothelial cell marker vWF. Only CKD-FCF expressed VCAM-1, a cell marker associated with inflammation. Incubation with TGF-β1 (0–10 ng/ml) induced a concentration dependent change in cell morphology, and upregulation of myofibroblast marker gene α-SMA expression alongside collagen 1α1, fibronectin, TGF-β1 and CTGF mRNA. These changes were blocked by the TGF-β1 receptor 1 antagonist SB431542 (5 μM). Conclusions FCF and CKD-FCF can be cultured via a simple method and represent a model for the investigation of the progression of fibrosis in feline CKD. The findings of this study suggest TGF-β1 may be involved in fibroblast-myofibroblast transition in feline CKD, as in other species.
Collapse
Affiliation(s)
- J S Lawson
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK.
| | - H M Syme
- Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Herts, AL9 7TA, UK
| | - C P D Wheeler-Jones
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - J Elliott
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| |
Collapse
|
42
|
Hewitson TD, Holt SG, Smith ER. Progression of Tubulointerstitial Fibrosis and the Chronic Kidney Disease Phenotype - Role of Risk Factors and Epigenetics. Front Pharmacol 2017; 8:520. [PMID: 28848437 PMCID: PMC5550676 DOI: 10.3389/fphar.2017.00520] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
Abstract
Although the kidney has capacity to repair after mild injury, ongoing or severe damage results in scarring (fibrosis) and an associated progressive loss of kidney function. However, despite its universal significance, evidence highlights a population based heterogeneity in the trajectory of chronic kidney disease (CKD) in these patients. To explain the heterogeneity of the CKD phenotype requires an understanding of the relevant risk factors for fibrosis. These factors include both the extrinsic nature of injury, and intrinsic factors such as age, gender, genetics, and perpetual activation of fibroblasts through priming. In many cases an additional level of regulation is provided by epigenetic mechanisms which integrate the various pro-fibrotic and anti-fibrotic triggers in fibrogenesis. In this review we therefore examine the various molecular and structural changes of fibrosis, and how they are influenced by extrinsic and intrinsic factors. Our aim is to provide a unifying hypothesis to help explain the transition from acute to CKD.
Collapse
Affiliation(s)
- Timothy D Hewitson
- Department of Nephrology, The Royal Melbourne Hospital, MelbourneVIC, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, MelbourneVIC, Australia
| | - Stephen G Holt
- Department of Nephrology, The Royal Melbourne Hospital, MelbourneVIC, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, MelbourneVIC, Australia
| | - Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital, MelbourneVIC, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, MelbourneVIC, Australia
| |
Collapse
|
43
|
Nlandu-Khodo S, Neelisetty S, Phillips M, Manolopoulou M, Bhave G, May L, Clark PE, Yang H, Fogo AB, Harris RC, Taketo MM, Lee E, Gewin LS. Blocking TGF- β and β-Catenin Epithelial Crosstalk Exacerbates CKD. J Am Soc Nephrol 2017; 28:3490-3503. [PMID: 28701516 DOI: 10.1681/asn.2016121351] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/08/2017] [Indexed: 11/03/2022] Open
Abstract
The TGF-β and Wnt/β-catenin pathways have important roles in modulating CKD, but how these growth factors affect the epithelial response to CKD is not well studied. TGF-β has strong profibrotic effects, but this pleiotropic factor has many different cellular effects depending on the target cell type. To investigate how TGF-β signaling in the proximal tubule, a key target and mediator of CKD, alters the response to CKD, we injured mice lacking the TGF-β type 2 receptor specifically in this epithelial segment. Compared with littermate controls, mice lacking the proximal tubular TGF-β receptor had significantly increased tubular injury and tubulointerstitial fibrosis in two different models of CKD. RNA sequencing indicated that deleting the TGF-β receptor in proximal tubule cells modulated many growth factor pathways, but Wnt/β-catenin signaling was the pathway most affected. We validated that deleting the proximal tubular TGF-β receptor impaired β-catenin activity in vitro and in vivo Genetically restoring β-catenin activity in proximal tubules lacking the TGF-β receptor dramatically improved the tubular response to CKD in mice. Deleting the TGF-β receptor alters many growth factors, and therefore, this ameliorated response may be a direct effect of β-catenin activity or an indirect effect of β-catenin interacting with other growth factors. In conclusion, blocking TGF-β and β-catenin crosstalk in proximal tubules exacerbates tubular injury in two models of CKD.
Collapse
Affiliation(s)
| | | | | | | | - Gautam Bhave
- Division of Nephrology, Department of Medicine and.,Departments of Cell and Developmental Biology
| | | | | | | | - Agnes B Fogo
- Division of Nephrology, Department of Medicine and.,Pathology, Microbiology and Immunology.,Pediatrics, and
| | - Raymond C Harris
- Division of Nephrology, Department of Medicine and.,Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee.,Departments of Medicine and
| | - M Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ethan Lee
- Departments of Cell and Developmental Biology
| | - Leslie S Gewin
- Division of Nephrology, Department of Medicine and .,Departments of Cell and Developmental Biology.,Research, Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee; and
| |
Collapse
|
44
|
Choi SE, Jeon N, Choi HY, Shin JI, Jeong HJ, Lim BJ. Lysyl oxidase‑like 2 is expressed in kidney tissue and is associated with the progression of tubulointerstitial fibrosis. Mol Med Rep 2017; 16:2477-2482. [PMID: 28677767 PMCID: PMC5548064 DOI: 10.3892/mmr.2017.6918] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/05/2017] [Indexed: 02/02/2023] Open
Abstract
Tubulointerstitial fibrosis is a common end point of chronic kidney diseases, and preventing its progression is key to avoiding renal failure. Transforming growth factor-β (TGF-β) and associated molecules promote tubulointerstitial fibrosis; however, effective therapies targeting these molecules have yet to be developed. Lysyl oxidase-like 2 (LOXL2), which is involved in invasive growth and metastasis of malignant neoplasms, has recently been reported to serve a key role in hepatic and pulmonary fibrosis. However, little is currently known regarding LOXL2 expression in the kidney and its involvement in tubulointerstitial fibrosis. The present study evaluated LOXL2 expression in human and mouse kidney tissues, as well as in cultured renal cells. LOXL2 protein expression was detected in glomerular capillary loops and tubular epithelial cells in human and mouse kidneys. Glomerular LOXL2 was localized to the cytoplasm of podocytes, as determined by double immunofluorescence microscopy using a podocyte marker (synaptopodin). This result was supported by western blot analysis, which demonstrated that LOXL2 protein expression is present in cultured human podocytes and HK-2 human proximal tubular cells. In addition, the mRNA and protein expression levels of LOXL2 were higher in a mouse model of tubulointerstitial fibrosis compared with in control mice. In addition, immunohistochemistry results demonstrated that LOXL2 is present in the fibrous interstitium and infiltrating mononuclear cells in a mouse model of tubulointerstitial fibrosis. The present study demonstrated that LOXL2 is expressed in compartments of renal tissue, where it appears to contribute to the progression of tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Sung-Eun Choi
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Nara Jeon
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hoon Young Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jae Il Shin
- Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyeon Joo Jeong
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Beom Jin Lim
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
45
|
A novel indole compound MA-35 attenuates renal fibrosis by inhibiting both TNF-α and TGF-β 1 pathways. Sci Rep 2017; 7:1884. [PMID: 28507324 PMCID: PMC5432497 DOI: 10.1038/s41598-017-01702-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/03/2017] [Indexed: 01/03/2023] Open
Abstract
Renal fibrosis is closely related to chronic inflammation and is under the control of epigenetic regulations. Because the signaling of transforming growth factor-β1 (TGF-β1) and tumor necrosis factor-α (TNF-α) play key roles in progression of renal fibrosis, dual blockade of TGF-β1 and TNF-α is desired as its therapeutic approach. Here we screened small molecules showing anti-TNF-α activity in the compound library of indole derivatives. 11 out of 41 indole derivatives inhibited the TNF-α effect. Among them, Mitochonic Acid 35 (MA-35), 5-(3, 5-dimethoxybenzyloxy)-3-indoleacetic acid, showed the potent effect. The anti-TNF-α activity was mediated by inhibiting IκB kinase phosphorylation, which attenuated the LPS/GaIN-induced hepatic inflammation in the mice. Additionally, MA-35 concurrently showed an anti-TGF-β1 effect by inhibiting Smad3 phosphorylation, resulting in the downregulation of TGF-β1-induced fibrotic gene expression. In unilateral ureter obstructed mouse kidney, which is a renal fibrosis model, MA-35 attenuated renal inflammation and fibrosis with the downregulation of inflammatory cytokines and fibrotic gene expressions. Furthermore, MA-35 inhibited TGF-β1-induced H3K4me1 histone modification of the fibrotic gene promoter, leading to a decrease in the fibrotic gene expression. MA-35 affects multiple signaling pathways involved in the fibrosis and may recover epigenetic modification; therefore, it could possibly be a novel therapeutic drug for fibrosis.
Collapse
|
46
|
Kidney development and perspectives for organ engineering. Cell Tissue Res 2017; 369:171-183. [DOI: 10.1007/s00441-017-2616-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/21/2017] [Indexed: 12/17/2022]
|
47
|
Zhu F, Bai X, Chen X. B lymphocytes in renal interstitial fibrosis. J Cell Commun Signal 2017; 11:213-218. [PMID: 28210941 DOI: 10.1007/s12079-017-0382-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/07/2017] [Indexed: 12/11/2022] Open
Abstract
Fibrosis is defined as an excessive deposition of extracellular matrix (ECM), which leads to the destruction of organ structure and impairment of organ function. Fibrosis occurs not only in kidney but also in lung, liver, heart, and skin. Common pathways of fibrosis are thought to exist. Renal interstitial fibrosis is a complex process that involves multiple molecular signaling and multiple cellular components, in which B cells appear to be one of the emerging important players. B cells may affect fibrosis through cytokine production and through interaction with other cells including fibroblasts, macrophages and T cells. This review summarizes recent research findings of B cells in fibrosis and provides an insight of how the future therapeutics of fibrosis could be developed from a B-cell point of view.
Collapse
Affiliation(s)
- Fengge Zhu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Xueyuan Bai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China.
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China.
| |
Collapse
|
48
|
Xavier S, Sahu RK, Landes SG, Yu J, Taylor RP, Ayyadevara S, Megyesi J, Stallcup WB, Duffield JS, Reis ES, Lambris JD, Portilla D. Pericytes and immune cells contribute to complement activation in tubulointerstitial fibrosis. Am J Physiol Renal Physiol 2017; 312:F516-F532. [PMID: 28052876 PMCID: PMC5374314 DOI: 10.1152/ajprenal.00604.2016] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/07/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022] Open
Abstract
We have examined the pathogenic role of increased complement expression and activation during kidney fibrosis. Here, we show that PDGFRβ-positive pericytes isolated from mice subjected to obstructive or folic acid injury secrete C1q. This was associated with increased production of proinflammatory cytokines, extracellular matrix components, collagens, and increased Wnt3a-mediated activation of Wnt/β-catenin signaling, which are hallmarks of myofibroblast activation. Real-time PCR, immunoblots, immunohistochemistry, and flow cytometry analysis performed in whole kidney tissue confirmed increased expression of C1q, C1r, and C1s as well as complement activation, which is measured as increased synthesis of C3 fragments predominantly in the interstitial compartment. Flow studies localized increased C1q expression to PDGFRβ-positive pericytes as well as to CD45-positive cells. Although deletion of C1qA did not prevent kidney fibrosis, global deletion of C3 reduced macrophage infiltration, reduced synthesis of C3 fragments, and reduced fibrosis. Clodronate mediated depletion of CD11bF4/80 high macrophages in UUO mice also reduced complement gene expression and reduced fibrosis. Our studies demonstrate local synthesis of complement by both PDGFRβ-positive pericytes and CD45-positive cells in kidney fibrosis. Inhibition of complement activation represents a novel therapeutic target to ameliorate fibrosis and progression of chronic kidney disease.
Collapse
Affiliation(s)
- Sandhya Xavier
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Ranjit K Sahu
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Susan G Landes
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Jing Yu
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Ronald P Taylor
- Department of Biochemistry, University of Virginia, Charlottesville, Virginia
| | | | - Judit Megyesi
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - William B Stallcup
- Sanford Burnham Prebys Medical Discovery Institute, Tumor Metastasis and Cancer Immunology Program, La Jolla, California
| | | | - Edimara S Reis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Didier Portilla
- Division of Nephrology, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia; .,Salem Veterans Affairs Medical Center, Salem, Virginia
| |
Collapse
|
49
|
Yang B, Chen S, Wu M, Zhang L, Ruan M, Chen X, Chen Z, Mei C, Mao Z. PHF14: an innate inhibitor against the progression of renal fibrosis following folic acid-induced kidney injury. Sci Rep 2017; 7:39888. [PMID: 28045076 PMCID: PMC5206671 DOI: 10.1038/srep39888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/29/2016] [Indexed: 02/06/2023] Open
Abstract
PHF14 is a newly identified regulator of mesenchyme growth in embryonic tissues. Previous studies have shown that phf14-null mutants die just after birth due to interstitial tissue hyperplasia in major organs, including the kidneys. The aim of this study was to investigate PHF14 function in renal fibrosis. By studying the chronic kidney injury mouse model, we found that PHF14 was upregulated in fibrotic kidneys after renal insults induced by folic acid administration. Compared with wild-type mice, PHF14-null mice showed more severe renal fibrosis after pro-fibrotic stimuli. Moreover, PHF14 in rat renal fibroblasts was upregulated by transforming growth factor-β (TGF-β) stimulation; while this upregulation was inhibited when smad3 phosphorylation was blocked. A chromatin immunoprecipitation (ChIP) assay further indicated that phospho-smad3 (p-smad3) acted as a transcription factor to enhance PHF14 expression. A lack of PHF14 expression enhanced collagen I and α-smooth muscle actin (α-SMA) synthesis induced by TGF-β in vitro. PHF14 was involved in inhibition of platelet-derived growth factor (PDGF) signaling overactivation by selectively repressing PDGF receptor-α (PDGFR-α) transcription. In summary, PHF14 expression was upregulated in fibrotic models in vivo and in vitro, and the TGF-β/smad3/PHF14 pathway acted as a self-limiting mechanism in the TGF-β-dominated renal pro-fibrotic process by suppressing PDGFR-α expression.
Collapse
Affiliation(s)
- Bo Yang
- Kidney Institute of CPLA, Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Sixiu Chen
- Kidney Institute of CPLA, Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Ming Wu
- Kidney Institute of CPLA, Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Lin Zhang
- School of Life Science and Technology, Shanghai Tech University, Shanghai 200031, People's Republic of China
| | - Mengna Ruan
- Kidney Institute of CPLA, Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Xujiao Chen
- Kidney Institute of CPLA, Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Zhengjun Chen
- School of Life Science and Technology, Shanghai Tech University, Shanghai 200031, People's Republic of China.,State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Changlin Mei
- Kidney Institute of CPLA, Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Zhiguo Mao
- Kidney Institute of CPLA, Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| |
Collapse
|
50
|
Gewin L, Zent R, Pozzi A. Progression of chronic kidney disease: too much cellular talk causes damage. Kidney Int 2016; 91:552-560. [PMID: 27773427 DOI: 10.1016/j.kint.2016.08.025] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/31/2016] [Accepted: 08/16/2016] [Indexed: 01/10/2023]
Abstract
Tubulointerstitial fibrosis, tubular atrophy, and peritubular capillary rarefaction are major hallmarks of chronic kidney disease. The tubulointerstitium consists of multiple cell components including tubular epithelial, mesenchymal (fibroblasts and pericytes), endothelial, and inflammatory cells. Crosstalk among these cell components is a key component in the pathogenesis of this complex disease. After severe or recurrent injury, the renal tubular epithelial cells undergo changes in structure and cell cycle that are accompanied by altered expression and production of cytokines. These cytokines contribute to the initiation of the fibrotic response by favoring activation of fibroblasts, recruitment of inflammatory cells, and loss of endothelial cells. This review focuses on how augmented growth factor and cytokine production induces epithelial crosstalk with cells in the interstitium to promote progressive tubulointerstitial fibrosis after renal injury.
Collapse
Affiliation(s)
- Leslie Gewin
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA; Department of Cancer Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA.
| | - Ambra Pozzi
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, Tennessee, USA; Veterans Affairs Medical Center, Nashville, Tennessee, USA; Department of Cancer Biology, Vanderbilt Medical Center, Nashville, Tennessee, USA; Department of Molecular Physiology and Biophysics, Vanderbilt Medical Center, Nashville, Tennessee, USA
| |
Collapse
|