1
|
Nørregaard R, Mutsaers HAM, Frøkiær J, Kwon TH. Obstructive nephropathy and molecular pathophysiology of renal interstitial fibrosis. Physiol Rev 2023; 103:2827-2872. [PMID: 37440209 PMCID: PMC10642920 DOI: 10.1152/physrev.00027.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023] Open
Abstract
The kidneys play a key role in maintaining total body homeostasis. The complexity of this task is reflected in the unique architecture of the organ. Ureteral obstruction greatly affects renal physiology by altering hemodynamics, changing glomerular filtration and renal metabolism, and inducing architectural malformations of the kidney parenchyma, most importantly renal fibrosis. Persisting pathological changes lead to chronic kidney disease, which currently affects ∼10% of the global population and is one of the major causes of death worldwide. Studies on the consequences of ureteral obstruction date back to the 1800s. Even today, experimental unilateral ureteral obstruction (UUO) remains the standard model for tubulointerstitial fibrosis. However, the model has certain limitations when it comes to studying tubular injury and repair, as well as a limited potential for human translation. Nevertheless, ureteral obstruction has provided the scientific community with a wealth of knowledge on renal (patho)physiology. With the introduction of advanced omics techniques, the classical UUO model has remained relevant to this day and has been instrumental in understanding renal fibrosis at the molecular, genomic, and cellular levels. This review details key concepts and recent advances in the understanding of obstructive nephropathy, highlighting the pathophysiological hallmarks responsible for the functional and architectural changes induced by ureteral obstruction, with a special emphasis on renal fibrosis.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jørgen Frøkiær
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
2
|
Cao R, Su W, Sheng J, Guo Y, Su J, Zhang C, Wang H, Tang Y, Chen L, Qiao R, Chen X, Huang X, Zhou Y, Zhu L, Bai Z, Zhang X, Gustafsson JA, Wan Q, Lan HY, Guan Y. Estrogen receptor β attenuates renal fibrosis by suppressing the transcriptional activity of Smad3. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166755. [PMID: 37196860 DOI: 10.1016/j.bbadis.2023.166755] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
Renal fibrosis (RF) is a common pathway leading to chronic kidney disease (CKD), which lacks effective treatment. While estrogen receptor beta (ERβ) is known to be present in the kidney, its role in RF remains unclear. The present study aimed to investigate the role and underlying mechanism of ERβ during RF progression in patients and animal models with CKD. We found that ERβ was highly expressed in the proximal tubular epithelial cells (PTECs) in healthy kidneys but its expression was largely lost in patients with immunoglobin A nephropathy (IgAN) and in mice with unilateral ureter obstruction (UUO) and subtotal nephrectomy (5/6Nx). ERβ deficiency markedly exacerbated, whereas ERβ activation by WAY200070 and DPN attenuated RF in both UUO and 5/6Nx mouse models, suggesting a protective role of ERβ in RF. In addition, ERβ activation inhibited TGF-β1/Smad3 signaling, while loss of renal ERβ was associated with overactivation of the TGF-β1/Smad3 pathway. Furthermore, deletion or pharmacological inhibition of Smad3 prevented the loss of ERβ and RF. Mechanistically, activation of ERβ competitively inhibited the association of Smad3 with the Smad-binding element, thereby downregulating the transcription of the fibrosis-related genes without altering Smad3 phosphorylation in vivo and in vitro. In conclusion, ERβ exerts a renoprotective role in CKD by blocking the Smad3 signaling pathway. Thus, ERβ may represent as a promising therapeutic agent for RF.
Collapse
Affiliation(s)
- Rong Cao
- Department of Nephrology, the First Affiliated Hospital of Shenzhen University, the Second People's Hospital of Shenzhen, Shenzhen 518035, China
| | - Wen Su
- Shenzhen University Health Science Center, Department of Pathology, Shenzhen University, Shenzhen 518071, China
| | - Jingyi Sheng
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210000, China
| | - Yanlin Guo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Jie Su
- Shenzhen University Health Science Center, Department of Pathology, Shenzhen University, Shenzhen 518071, China
| | - Cong Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Honglian Wang
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Research Center for Integrative Medicine, the Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yizhe Tang
- The First Affiliated Hospital of Shenzhen University, Health Science Center, China; The Second People's Hospital of Shenzhen, Institute of Translational Medicine, Medical Research Center, China
| | - Lei Chen
- Shenzhen University Health Science Center, Department of Pathology, Shenzhen University, Shenzhen 518071, China
| | - Rongfang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Xiaocong Chen
- Shenzhen University Health Science Center, Department of Pathology, Shenzhen University, Shenzhen 518071, China
| | - Xiaoru Huang
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou 510080, China
| | - Yunfeng Zhou
- Department of Physiology at the Basic Medical College, Shenzhen University Health Science Center, Shenzhen 518071, China
| | - Lizhen Zhu
- Shenzhen University Health Science Center, Department of Pathology, Shenzhen University, Shenzhen 518071, China
| | - Zirui Bai
- Shenzhen University Health Science Center, Department of Pathology, Shenzhen University, Shenzhen 518071, China
| | - Xiaoyan Zhang
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Jan-Ake Gustafsson
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden; Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, USA
| | - Qijun Wan
- Department of Nephrology, the First Affiliated Hospital of Shenzhen University, the Second People's Hospital of Shenzhen, Shenzhen 518035, China.
| | - Hui-Yao Lan
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning, China.
| |
Collapse
|
3
|
Kocic G, Gajic M, Tomovic K, Hadzi-Djokic J, Anderluh M, Smelcerovic A. Purine adducts as a presumable missing link for aristolochic acid nephropathy-related cellular energy crisis, potential anti-fibrotic prevention and treatment. Br J Pharmacol 2021; 178:4411-4427. [PMID: 34235731 DOI: 10.1111/bph.15618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 06/15/2021] [Accepted: 06/28/2021] [Indexed: 12/20/2022] Open
Abstract
Aristolochic acid nephropathy is a progressive exposome-induced disease characterized by tubular atrophy and fibrosis culminating in end-stage renal disease and malignancies. The molecular mechanisms of the energy crisis as a putative cause of fibrosis have not yet been elucidated. In light of the fact that aristolochic acid forms DNA and RNA adducts by covalent binding of aristolochic acid metabolites to exocyclic amino groups of (deoxy)adenosine and (deoxy)guanosine, we hypothesize here that similar aristolochic acid adducts may exist with other purine-containing molecules. We also provide new insights into the aristolochic acid-induced energy crisis and presumably a link between already known mechanisms. In addition, an overview of potential targets in fibrosis treatment is provided, which is followed by recommendations on possible preventive measures that could be taken to at least postpone or partially alleviate aristolochic acid nephropathy.
Collapse
Affiliation(s)
- Gordana Kocic
- Department of Biochemistry, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Mihajlo Gajic
- Department of Pharmacy, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Katarina Tomovic
- Department of Pharmacy, Faculty of Medicine, University of Nis, Nis, Serbia
| | | | - Marko Anderluh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Andrija Smelcerovic
- Department of Chemistry, Faculty of Medicine, University of Nis, Nis, Serbia
| |
Collapse
|
4
|
Higgins CE, Tang J, Higgins SP, Gifford CC, Mian BM, Jones DM, Zhang W, Costello A, Conti DJ, Samarakoon R, Higgins PJ. The Genomic Response to TGF-β1 Dictates Failed Repair and Progression of Fibrotic Disease in the Obstructed Kidney. Front Cell Dev Biol 2021; 9:678524. [PMID: 34277620 PMCID: PMC8284093 DOI: 10.3389/fcell.2021.678524] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Tubulointerstitial fibrosis is a common and diagnostic hallmark of a spectrum of chronic renal disorders. While the etiology varies as to the causative nature of the underlying pathology, persistent TGF-β1 signaling drives the relentless progression of renal fibrotic disease. TGF-β1 orchestrates the multifaceted program of kidney fibrogenesis involving proximal tubular dysfunction, failed epithelial recovery or re-differentiation, capillary collapse and subsequent interstitial fibrosis eventually leading to chronic and ultimately end-stage disease. An increasing complement of non-canonical elements function as co-factors in TGF-β1 signaling. p53 is a particularly prominent transcriptional co-regulator of several TGF-β1 fibrotic-response genes by complexing with TGF-β1 receptor-activated SMADs. This cooperative p53/TGF-β1 genomic cluster includes genes involved in cellular proliferative control, survival, apoptosis, senescence, and ECM remodeling. While the molecular basis for this co-dependency remains to be determined, a subset of TGF-β1-regulated genes possess both p53- and SMAD-binding motifs. Increases in p53 expression and phosphorylation, moreover, are evident in various forms of renal injury as well as kidney allograft rejection. Targeted reduction of p53 levels by pharmacologic and genetic approaches attenuates expression of the involved genes and mitigates the fibrotic response confirming a key role for p53 in renal disorders. This review focuses on mechanisms underlying TGF-β1-induced renal fibrosis largely in the context of ureteral obstruction, which mimics the pathophysiology of pediatric unilateral ureteropelvic junction obstruction, and the role of p53 as a transcriptional regulator within the TGF-β1 repertoire of fibrosis-promoting genes.
Collapse
Affiliation(s)
- Craig E. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Jiaqi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Stephen P. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Cody C. Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Badar M. Mian
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - David M. Jones
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, NY, United States
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Angelica Costello
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - David J. Conti
- Division of Transplantation Surgery, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Paul J. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| |
Collapse
|
5
|
Mukherjee M, Fogarty E, Janga M, Surendran K. Notch Signaling in Kidney Development, Maintenance, and Disease. Biomolecules 2019; 9:E692. [PMID: 31690016 PMCID: PMC6920979 DOI: 10.3390/biom9110692] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/31/2019] [Accepted: 11/02/2019] [Indexed: 02/06/2023] Open
Abstract
Kidney development involves formation of nephrons intricately aligned with the vasculature and connected to a branched network of collecting ducts. Notch signaling plays multiple roles during kidney development involving the formation of nephrons composed of diverse epithelial cell types arranged into tubular segments, all the while maintaining a nephron progenitor niche. Here, we review the roles of Notch signaling identified from rodent kidney development and injury studies, while discussing human kidney diseases associated with aberrant Notch signaling. We also review Notch signaling requirement in maintenance of mature kidney epithelial cell states and speculate that Notch activity regulation mediates certain renal physiologic adaptations.
Collapse
Affiliation(s)
- Malini Mukherjee
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA.
| | - Eric Fogarty
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA.
| | - Madhusudhana Janga
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA.
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA.
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA.
| |
Collapse
|
6
|
Chen X, Wu Y, Diao Z, Han X, Li D, Ruan X, Liu W. C1q/tumor necrosis factor‐related protein‐3 improves renal fibrosis via inhibiting notch signaling pathways. J Cell Physiol 2019; 234:22352-22364. [PMID: 31074042 DOI: 10.1002/jcp.28801] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/20/2019] [Accepted: 04/24/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Xinpan Chen
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases Capital Medical University Beijing China
| | - Yiru Wu
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases Capital Medical University Beijing China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases Capital Medical University Beijing China
| | - Xue Han
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases Capital Medical University Beijing China
| | - Dishan Li
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases Capital Medical University Beijing China
| | - Xiongzhong Ruan
- Department of Nephrology, John Moorhead Research Laboratory, University College London Medical School, Royal Free Campus University College London London United Kingdom
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases Capital Medical University Beijing China
| |
Collapse
|
7
|
Hammad FT, Salam SA, Nemmar A, Ali M, Lubbad L. The Effect of Arabic Gum on Renal Function in Reversible Unilateral Ureteric Obstruction. Biomolecules 2019; 9:biom9010025. [PMID: 30641998 PMCID: PMC6359443 DOI: 10.3390/biom9010025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 02/07/2023] Open
Abstract
Arabic gum (AG) has antioxidant and anti-inflammatory properties. However, the effect of AG in ureteric obstruction (UO) has not been investigated yet. Male rats underwent reversible left unilateral UO (UUO) for 72 h. Group AG-1 (n = 12) received AG 15 g/kg/day dissolved in drinking water starting seven days before and continuing throughout the period of the UUO, whereas group Vx-1 (n = 8) had only water. Group AG-2 (n = 12) and Vx-2 (n = 8) had similar protocols as AG-1 and Vx-1, respectively, but underwent terminal experiments to measure renal functions, six days post-UUO reversal. Arabic gum significantly attenuated the UUO-induced increase in the tissue level of malonedialdehyde and superoxide dismutase and the rise in the gene expression of TNF-α, TGF-β1, and p53 in AG-1 compared to Vx-1. It also attenuated the severity of tubular dilatation. However, AG did not affect the alterations in the renal blood flow or glomerular filtration rate. The fractional sodium excretion was lower in AG-2 but did not reach statistical significance (0.40 ± 0.11 vs 0.74 ± 0.12, p = 0.07). AG attenuated the UUO-induced rise in oxidative stress markers and proinflammatory and profibrotic cytokines and the degree of renal tubular dilatation, indicating a protective effect in obstructive nephropathy.
Collapse
Affiliation(s)
- Fayez T Hammad
- Department of Surgery, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain 17666, UAE.
| | - Suhail Al Salam
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain 17666, UAE.
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain 17666, UAE.
| | - Mahmoud Ali
- Department of Pharmacology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain 17666, UAE.
| | - Loay Lubbad
- Department of Surgery, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain 17666, UAE.
| |
Collapse
|
8
|
Ma N, Kato T, Isogai T, Gu Y, Yamashita T. The Potential Effects of Taurine in Mitigation of Radiation Nephropathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1155:497-505. [PMID: 31468426 DOI: 10.1007/978-981-13-8023-5_46] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Taurine (2-aminoethanesulfonic acid) is a sulfur-containing organic acid possessing several important effects, including antioxidant and anti-inflammatory ones. Exposure to ionizing radiation generates free radicals and reactive oxygen species (ROS) in irradiated cells, and free radical generation leads to oxidative stress. It is known that radiation nephropathy includes an inflammation-based process in which ROS and cytokines are responsible. Different doses of explored radiation can cause apoptosis, inflammation and a profound oxidative stress in kidneys. Oxidative stress is involved in renal injury after exposure to both ionizing radiation and inflammation. In this review, we describe the protective effect of taurine against several kidney diseases and the potential effects of taurine in the mitigation of radiation nephropathy. We also report that X-irradiation decreased the expression of taurine and TauT in the kidney. Taurine administration suppressed the decrease in the expression of taurine and TauT in the kidney after radiation exposure. Taurine might contribute to the mitigation of kidney injury induced by radiation.
Collapse
Affiliation(s)
- Ning Ma
- Division of Health Science, Graduate School of Health Science, Suzuka University, Suzuka, Mie, Japan
| | - Toshihiro Kato
- Department of Rehabilitation, Suzuka Kaisei Hospital, Suzuka, Japan
| | - Tamami Isogai
- Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Japan
| | - Yeunhwa Gu
- Faculty of Health Science, Junshin Gakuen University, Fukuoka, Japan
| | - Takenori Yamashita
- Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Japan. .,Division of Health Science, Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Mie, Japan.
| |
Collapse
|
9
|
Ma H, Lee S, Yang Y, Bedi P, Chou SY. Pentoxifylline protects against loss of function and renal interstitial fibrosis in chronic experimental partial ureteral obstruction. PATHOPHYSIOLOGY 2018; 25:419-425. [DOI: 10.1016/j.pathophys.2018.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/14/2018] [Accepted: 08/22/2018] [Indexed: 12/21/2022] Open
|
10
|
5-aminoisoquinoline improves renal function and fibrosis during recovery phase of cisplatin-induced acute kidney injury in rats. Biosci Rep 2018; 38:BSR20171313. [PMID: 29599129 PMCID: PMC5920139 DOI: 10.1042/bsr20171313] [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] [Received: 09/29/2017] [Revised: 03/22/2018] [Accepted: 03/29/2018] [Indexed: 01/23/2023] Open
Abstract
The aim of the present study is to analyze the effects of 5-aminoisoquinoline (5-AIQ), a poly(ADP-ribose) polymerase-1 (PARP1) inhibitor, over renal dysfunction and fibrosis during recovery phase of cisplatin (CisPt)-induced acute kidney injury (AKI) in rats. Male Wistar rats were distributed in three groups (n=8 each group): control, CisPt, and CisPt + 5-AIQ. Control and CisPt groups received a subcutaneous injection of either saline or 7 mg/kg CisPt, respectively. CisPt + 5-AIQ group received two intraperitoneal injections of 10 mg/kg 5-AIQ 2 h before and 24 h after CisPt treatment. Thirteen days after the treatment, rats were housed in metabolic cages and 24-h urine collection was made. At day 14, CisPt-treated rats showed increased diuresis, N-acetyl-β-d-glucosaminidase (NAG) excretion, glucosuria and sodium fractional excretion (NaFE), and decreased creatinine clearance (CrCl). 5-AIQ significantly increased CrCl and decreased NAG excretion, glucosuria, and NaFE. In plasma, CisPt increased sodium, urea, and creatinine concentrations, while 5-AIQ treatment decreased these variables to the levels of control group. 5-AIQ completely prevented the body weight loss evoked by CisPt treatment. CisPt also induced an increased renal expression of PAR polymer, α-smooth muscle actin (α-SMA), transforming growth factor-β1 (TGF-β1), and collagen-IV. These variables were decreased in CisPt + 5-AIQ group. Tubular lesions and renal fibrosis were also decreased by 5-AIQ treatment. We conclude that inhibition of PARP1 with 5-AIQ can attenuate long-term nephrotoxic effects associated with the CisPt treatment, preventing renal dysfunction and body weight decrease and ameliorating tubular lesions and collagen deposition.
Collapse
|
11
|
Chen H, Yang T, Wang MC, Chen DQ, Yang Y, Zhao YY. Novel RAS inhibitor 25-O-methylalisol F attenuates epithelial-to-mesenchymal transition and tubulo-interstitial fibrosis by selectively inhibiting TGF-β-mediated Smad3 phosphorylation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 42:207-218. [PMID: 29655688 DOI: 10.1016/j.phymed.2018.03.034] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/31/2018] [Accepted: 03/17/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Tubulo-interstitial fibrosis (TIF) is the common pathway in the chronic kidney disease (CKD). Epithelial-to-mesenchymal transition (EMT) is a major contributor to the TIF by the increased myofibroblasts. Renin-angiotensin system (RAS) is critical mediator on EMT in progressive CKD. Angiotensin II (ANG) mediates EMT and causes TIF by stimulating transforming growth factor-β1 (TGF-β1). RAS activation could further activate TGF-β1. Inhibition of the RAS is one of the most powerful therapies for progressive CKD. 25-O-methylalisol F (MAF) is a new tetracyclic triterpenoid compound isolated from the Alismatis rhizoma, which is extensively used for anti-hypertensive, diuretic and anti-hyperlipidemic effects. METHODS Inhibitory effect of MAF on EMT is investigated in both TGF-β1- and ANG-induced tubular epithelial cells (NRK-52E) and fibroblasts (NRK-49F). Western blot analysis, qRT-PCR, siRNA, immunofluorescence staining and co-immunoprecipitation techniques were used to evaluate the inhibition of MAF on EMT and further revealed the intervention effects on RAS, TGF-β/Smad and Wnt/β-catenin pathways. RESULTS MAF treatment significantly inhibited TGF-β1 and ANG-induced expressions of collagen I, fibronectin, α-SMA, vimentin and E-cadherin at both mRNA and protein levels in the NRK-52E and NRK-49F cells. The action mechanism revealed that MAF significantly ameliorated upregulation of angiotensinogen, renin, ACE and AT1R expressions. Further, MAF attenuated upregulation of Smad3 phosphorylation and downregulation of Smad7, but did not affect the phosphorylation of Smad2, PI3K, ERK1/2 and p38 expressions and Smad4 expression in NRK-52E cells. Co-immunoprecipitation analysis indicated that MAF selectively blocked the combination of Smad3 with TGFβRI and Smad3 with SARA without interfering with the Smad2, TGFβRI and SARA interaction. Additionally, MAF suppressed the expressions of Wnt1 and β-catenin as well as its downstream target Snail1, Twist, MMP-7, PAI-1 and FSP1 expressions in NRK-52E cells. CONCLUSIONS MAF simultaneously targeted multiple RAS components and it was a novel RAS inhibitor. MAF inhibited EMT by Smad3-specific signaling in the TGF-β/Smad-dependent pathway and Wnt/β-catenin pathway. MAF has an important effect on crosstalk between the TGF-β/Smad and Wnt/β-catenin pathway in EMT process by activation of RAS.
Collapse
Affiliation(s)
- Hua Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Tian Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Min-Chang Wang
- Xi'an Modern Chemistry Institute, Xi'an, Shaanxi 710065, China
| | - Dan-Qian Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Ying-Yong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
12
|
Is hydrogen sulfide a potential novel therapy to prevent renal damage during ureteral obstruction? Nitric Oxide 2018; 73:15-21. [DOI: 10.1016/j.niox.2017.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/12/2017] [Accepted: 12/17/2017] [Indexed: 12/28/2022]
|
13
|
Jackson L, Woodward M, Coward RJ. The molecular biology of pelvi-ureteric junction obstruction. Pediatr Nephrol 2018; 33:553-571. [PMID: 28286898 PMCID: PMC5859056 DOI: 10.1007/s00467-017-3629-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/17/2022]
Abstract
Over recent years routine ultrasound scanning has identified increasing numbers of neonates as having hydronephrosis and pelvi-ureteric junction obstruction (PUJO). This patient group presents a diagnostic and management challenge for paediatric nephrologists and urologists. In this review we consider the known molecular mechanisms underpinning PUJO and review the potential of utilising this information to develop novel therapeutics and diagnostic biomarkers to improve the care of children with this disorder.
Collapse
Affiliation(s)
- Laura Jackson
- Bristol Renal Group, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK. .,Bristol Royal Hospital for Children, Bristol, UK.
| | - Mark Woodward
- 0000 0004 0399 4960grid.415172.4Bristol Royal Hospital for Children, Bristol, UK
| | - Richard J. Coward
- 0000 0004 1936 7603grid.5337.2Bristol Renal Group, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY UK ,0000 0004 0399 4960grid.415172.4Bristol Royal Hospital for Children, Bristol, UK
| |
Collapse
|
14
|
Chemical chaperon 4-phenylbutyrate protects against the endoplasmic reticulum stress-mediated renal fibrosis in vivo and in vitro. Oncotarget 2017; 7:22116-27. [PMID: 26959118 PMCID: PMC5008348 DOI: 10.18632/oncotarget.7904] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/21/2016] [Indexed: 12/16/2022] Open
Abstract
Renal tubulointerstitial fibrosis is the common and final pathologic change of kidney in end-stage renal disease. Interesting, endoplasmic reticulum (ER) stress is known to contribute to the pathophysiological mechanisms during the development of renal fibrosis. Here, we investigated the effects of chemical chaperon sodium 4-phenylbutyrate (4-PBA) on renal fibrosis in vivo and in vitro. In a rat unilateral ureteral obstruction (UUO) model, 4-PBA mimicked endogenous ER chaperon in the kidneys and significantly reduced glucose regulated protein 78 (GRP78), CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP), activating transcription factor 4 (ATF4), and phosphorylated JNK protein expressions as well as restored spliced X-box-binding protein 1 (XBP1) expressions in the kidneys of UUO rats. 4-PBA also attenuated the increases of α-smooth muscle actin (α-SMA), connective tissue growth factor (CTGF) protein expressions, tubulointerstitial fibrosis, and apoptosis in the kidneys of UUO rats. Moreover, transforming growth factor (TGF)-β markedly increased ER stress-associated molecules, profibrotic factors, and apoptotic markers in the renal tubular cells (NRK-52E), all of which could be significantly counteracted by 4-PBA treatment. 4-PBA also diminished TGF-β-increased CTGF promoter activity and CTGF mRNA expression in NRK-52E cells. Taken together, our results indicated that 4-PBA acts as an ER chaperone to ameliorate ER stress-induced renal tubular cell apoptosis and renal fibrosis.
Collapse
|
15
|
McGaraughty S, Davis-Taber RA, Zhu CZ, Cole TB, Nikkel AL, Chhaya M, Doyle KJ, Olson LM, Preston GM, Grinnell CM, Salte KM, Giamis AM, Luo Y, Sun V, Goodearl AD, Gopalakrishnan M, Lacy SE. Targeting Anti-TGF- β Therapy to Fibrotic Kidneys with a Dual Specificity Antibody Approach. J Am Soc Nephrol 2017; 28:3616-3626. [PMID: 28827403 PMCID: PMC5698069 DOI: 10.1681/asn.2017010013] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/03/2017] [Indexed: 01/06/2023] Open
Abstract
Targeted delivery of a therapeutic agent to a site of pathology to ameliorate disease while limiting exposure at undesired tissues is an aspirational treatment scenario. Targeting diseased kidneys for pharmacologic treatment has had limited success. We designed an approach to target an extracellular matrix protein, the fibronectin extra domain A isoform (FnEDA), which is relatively restricted in distribution to sites of tissue injury. In a mouse unilateral ureteral obstruction (UUO) model of renal fibrosis, injury induced significant upregulation of FnEDA in the obstructed kidney. Using dual variable domain Ig (DVD-Ig) technology, we constructed a molecule with a moiety to target FnEDA and a second moiety to neutralize TGF-β After systemic injection of the bispecific TGF-β + FnEDA DVD-Ig or an FnEDA mAb, chemiluminescent detection and imaging with whole-body single-photon emission computed tomography (SPECT) revealed significantly higher levels of each molecule in the obstructed kidney than in the nonobstructed kidney, the ipsilateral kidney of sham animals, and other tissues. In comparison, a systemically administered TGF-β mAb accumulated at lower concentrations in the obstructed kidney and exhibited a more diffuse whole-body distribution. Systemic administration of the bispecific DVD-Ig or the TGF-β mAb (1-10 mg/kg) but not the FnEDA mAb attenuated the injury-induced collagen deposition detected by immunohistochemistry and elevation in Col1a1, FnEDA, and TIMP1 mRNA expression in the obstructed kidney. Overall, systemic delivery of a bispecific molecule targeting an extracellular matrix protein and delivering a TGF-β mAb resulted in a relatively focal uptake in the fibrotic kidney and reduced renal fibrosis.
Collapse
Affiliation(s)
| | | | - Chang Z Zhu
- AbbVie Discovery Lake County, North Chicago, Illinois; and
| | - Todd B Cole
- AbbVie Discovery Lake County, North Chicago, Illinois; and
| | | | - Meha Chhaya
- AbbVie Bioresearch Center, Worcester, Massachusetts
| | - Kelly J Doyle
- AbbVie Discovery Lake County, North Chicago, Illinois; and
| | - Lauren M Olson
- AbbVie Discovery Lake County, North Chicago, Illinois; and
| | | | | | | | | | - Yanping Luo
- AbbVie Discovery Lake County, North Chicago, Illinois; and
| | - Victor Sun
- AbbVie Bioresearch Center, Worcester, Massachusetts
| | | | | | - Susan E Lacy
- AbbVie Bioresearch Center, Worcester, Massachusetts
| |
Collapse
|
16
|
Matheus LHG, Simão GM, Amaral TA, Brito RBO, Malta CS, Matos YST, Santana AC, Rodrigues GGC, Albejante MC, Bach EE, Dalboni MA, Camacho CP, Dellê H. Indoleamine 2, 3-dioxygenase (IDO) increases during renal fibrogenesis and its inhibition potentiates TGF-β 1-induced epithelial to mesenchymal transition. BMC Nephrol 2017; 18:287. [PMID: 28877670 PMCID: PMC5585959 DOI: 10.1186/s12882-017-0702-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/22/2017] [Indexed: 01/01/2023] Open
Abstract
Background Indoleamine 2, 3-dioxygenase (IDO) is an immunomodulatory molecule that has been implicated in several biological processes. Although IDO has been linked with some renal diseases, its role in renal fibrosis is still unclear. Because IDO may be modulated by TGF-β1, a potent fibrogenic molecule, we hypothesized that IDO could be involved in renal fibrosis, especially acting in the TGF-β1-induced tubular epithelial-mesenchymal transition (EMT). We analyzed the IDO expression and activity in a model of renal fibrogenesis, and the effect of the IDO inhibitor 1-methyl-tryptophan (MT) on TGF-β1-induced EMT using tubular cell culture. Methods Male Wistar rats where submited to 7 days of UUO. Non-obstructed kidneys (CL) and kidneys from SHAM rats were used as controls. Masson’s Tricrome and macrophages counting were used to chatacterize the tissue fibrosis. The EMT was analysed though immunohistochemistry and qRT-PCR. Immunohistochemestry in tissue has used to show IDO expression. MDCK cells were incubated with TGF- β1 to analyse IDO expression. Additionally, effects of TGF- β1 and the inhibition of IDO over the EMT process was acessed by immunoessays and scrath wound essay. Results IDO was markedly expressed in cortical and medular tubules of the UUO kidneys. Similarly to the immunolocalizaton of TGF- β1, accompanied by loss of e-cadherin expression and an increase of mesenchymal markers. Results in vitro with MDCK cells, showed that IDO was increased after stimulus with TGF-β1, and treatment with MT potentiated its expression. MDCK stimulated with TGF-β1 had higher migratory activity (scratch-wound assay), which was exacerbated by MT treatment. Conclusions IDO is constitutively expressed in tubular cells and increases during renal fibrogenesis. Although IDO is induced by TGF-β1 in tubular cells, its chemical inhibitor acts as a profibrotic agent.
Collapse
Affiliation(s)
- Luiz Henrique Gomes Matheus
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Gislene Mendes Simão
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Taíssa Altieri Amaral
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Rodrigo Barbosa Oliveira Brito
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Camila Soares Malta
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Yves Silva Teles Matos
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Alexandre Chagas Santana
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Gabriela Gomes Cardoso Rodrigues
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Maria Clara Albejante
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Erna Elisabeth Bach
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Maria Aparecida Dalboni
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Cleber Pinto Camacho
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil
| | - Humberto Dellê
- Postgraduate Program in Medicine, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235, 2º subsolo, São Paulo, 01504-001, São Paulo, Brazil.
| |
Collapse
|
17
|
Itano S, Satoh M, Kadoya H, Sogawa Y, Uchida A, Sasaki T, Kashihara N. Colchicine attenuates renal fibrosis in a murine unilateral ureteral obstruction model. Mol Med Rep 2017; 15:4169-4175. [DOI: 10.3892/mmr.2017.6539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 02/23/2017] [Indexed: 11/05/2022] Open
|
18
|
Nagata N, Iwanari H, Kumagai H, Kusano-Arai O, Ikeda Y, Aritake K, Hamakubo T, Urade Y. Generation and characterization of an antagonistic monoclonal antibody against an extracellular domain of mouse DP2 (CRTH2/GPR44) receptors for prostaglandin D2. PLoS One 2017; 12:e0175452. [PMID: 28394950 PMCID: PMC5386288 DOI: 10.1371/journal.pone.0175452] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 03/27/2017] [Indexed: 01/09/2023] Open
Abstract
Prostaglandin D2 (PGD2) is a lipid mediator involved in sleep regulation and inflammation. PGD2 interacts with 2 types of G protein-coupled receptors, DP1 and DP2/CRTH2 (chemoattractant receptor homologous molecule expressed on T helper type 2 cells)/GPR44 to show a variety of biological effects. DP1 activation leads to Gs-mediated elevation of the intracellular cAMP level, whereas activation of DP2 decreases this level via the Gi pathway; and it also induces G protein-independent, arrestin-mediated cellular responses. Activation of DP2 by PGD2 causes the progression of inflammation via the recruitment of lymphocytes by enhancing the production of Th2-cytokines. Here we developed monoclonal antibodies (MAbs) against the extracellular domain of mouse DP2 by immunization of DP2-null mutant mice with DP2-overexpressing BAF3, murine interleukin-3 dependent pro-B cells, to reduce the generation of antibodies against the host cells by immunization of mice. Moreover, we immunized DP2-KO mice to prevent immunological tolerance to mDP2 protein. After cell ELISA, immunocytochemical, and Western blot analyses, we successfully obtained a novel monoclonal antibody, MAb-1D8, that specifically recognized native mouse DP2, but neither human DP2 nor denatured mouse DP2, by binding to a particular 3D receptor conformation formed by the N-terminus and extracellular loop 1, 2, and 3 of DP2. This antibody inhibited the binding of 0.5 nM [3H]PGD2 to mouse DP2 (IC50 = 46.3 ± 18.6 nM), showed antagonistic activity toward 15(R)-15-methyl PGD2-induced inhibition of 300 nM forskolin-activated cAMP production (IC50 = 16.9 ± 2.6 nM), and gave positive results for immunohistochemical staining of DP2-expressing CD4+ Th2 lymphocytes that had accumulated in the kidney of unilateral ureteral obstruction model mice. This monoclonal antibody will be very useful for in vitro and in vivo studies on DP2-mediated diseases.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibody Specificity
- CD4-Positive T-Lymphocytes/metabolism
- CHO Cells
- COS Cells
- Cricetulus
- Cyclic AMP/metabolism
- Disease Models, Animal
- Epitope Mapping
- HEK293 Cells
- Humans
- Hybridomas/metabolism
- Immunization
- Immunohistochemistry
- Kidney/metabolism
- Kidney/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Precursor Cells, B-Lymphoid/immunology
- Prostaglandin D2/analogs & derivatives
- Prostaglandin D2/antagonists & inhibitors
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Prostaglandin/genetics
- Receptors, Prostaglandin/immunology
- Ureteral Obstruction/immunology
- Ureteral Obstruction/metabolism
- Ureteral Obstruction/pathology
- beta-Arrestins/metabolism
Collapse
Affiliation(s)
- Nanae Nagata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Furuedai, Suita, Osaka, Japan
- * E-mail: (YU); (NN)
| | - Hiroko Iwanari
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hidetoshi Kumagai
- Department of Advanced Clinical Science and Therapeutics, The University of Tokyo, Tokyo, Japan
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Osamu Kusano-Arai
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yuichi Ikeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kosuke Aritake
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Furuedai, Suita, Osaka, Japan
| | - Takao Hamakubo
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Urade
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Furuedai, Suita, Osaka, Japan
- * E-mail: (YU); (NN)
| |
Collapse
|
19
|
Pap D, Sziksz E, Kiss Z, Rokonay R, Veres-Székely A, Lippai R, Takács IM, Kis É, Fekete A, Reusz G, Szabó AJ, Vannay A. Microarray Analysis Reveals Increased Expression of Matrix Metalloproteases and Cytokines of Interleukin-20 Subfamily in the Kidneys of Neonate Rats Underwent Unilateral Ureteral Obstruction: A Potential Role of IL-24 in the Regulation of Inflammation and Tissue Remodeling. Kidney Blood Press Res 2017; 42:16-32. [PMID: 28253513 DOI: 10.1159/000464317] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 12/21/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Congenital obstructive nephropathy (CON) is the main cause of pediatric chronic kidney diseases leading to renal fibrosis. High morbidity and limited treatment opportunities of CON urge the better understanding of the underlying molecular mechanisms. METHODS To identify the differentially expressed genes, microarray analysis was performed on the kidney samples of neonatal rats underwent unilateral ureteral obstruction (UUO). Microarray results were then validated by real-time RT-PCR and bioinformatics analysis was carried out to identify the relevant genes, functional groups and pathways involved in the pathomechanism of CON. Renal expression of matrix metalloproteinase (MMP)-12 and interleukin (IL)-24 were evaluated by real-time RT-PCR, flow cytometry and immunohistochemical analysis. Effect of the main profibrotic factors on the expression of MMP-12 and IL-24 was investigated on HK-2 and HEK-293 cell lines. Finally, the effect of IL-24 treatment on the expression of pro-inflammatory cytokines and MMPs were tested in vitro. RESULTS Microarray analysis revealed 880 transcripts showing >2.0-fold change following UUO, enriched mainly in immune response related processes. The most up-regulated genes were MMPs and members of IL-20 cytokine subfamily, including MMP-3, MMP-7, MMP-12, IL-19 and IL-24. We found that while TGF-β treatment inhibits the expression of MMP-12 and IL-24, H2O2 or PDGF-B treatment induce the epithelial expression of MMP-12. We demonstrated that IL-24 treatment decreases the expression of IL-6 and MMP-3 in the renal epithelial cells. CONCLUSIONS This study provides an extensive view of UUO induced changes in the gene expression profile of the developing kidney and describes novel molecules, which may play significant role in the pathomechanism of CON.
Collapse
Affiliation(s)
- Domonkos Pap
- MTA-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Erna Sziksz
- MTA-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Zoltán Kiss
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Réka Rokonay
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Apor Veres-Székely
- MTA-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Rita Lippai
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | | | - Éva Kis
- MTA-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Andrea Fekete
- MTA-SE, Lendület Diabetes Research Group, Budapest, Hungary
| | - György Reusz
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Attila J Szabó
- MTA-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Adam Vannay
- MTA-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| |
Collapse
|
20
|
Ledo N, Susztak K, Palmer MB. Cell Phenotype Transitions in Renal Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2016. [DOI: 10.1007/s40139-016-0098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
Angiopoietin-like protein 2 increases renal fibrosis by accelerating transforming growth factor-β signaling in chronic kidney disease. Kidney Int 2016; 89:327-41. [DOI: 10.1016/j.kint.2015.12.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 09/16/2015] [Accepted: 10/07/2015] [Indexed: 01/08/2023]
|
22
|
Gregorini M, Corradetti V, Rocca C, Pattonieri EF, Valsania T, Milanesi S, Serpieri N, Bedino G, Esposito P, Libetta C, Avanzini MA, Mantelli M, Ingo D, Peressini S, Albertini R, Dal Canton A, Rampino T. Mesenchymal Stromal Cells Prevent Renal Fibrosis in a Rat Model of Unilateral Ureteral Obstruction by Suppressing the Renin-Angiotensin System via HuR. PLoS One 2016; 11:e0148542. [PMID: 26866372 PMCID: PMC4750962 DOI: 10.1371/journal.pone.0148542] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
We studied Mesenchymal Stromal Cells (MSC) effects in experimental Unilateral Ureteral Obstruction (UUO), a fibrogenic renal disease. Rats were divided in 5 groups: sham, UUO, MSC treated-UUO, ACEi treated-UUO, MSC+ACEi treated- UUO. Data were collected at 1, 7, 21 days. UUO induced monocyte renal infiltration, tubular cell apoptosis, tubular atrophy, interstitial fibrosis and overexpression of TGFβ, Renin mRNA (RENmRNA), increase of Renin, Angiotensin II (AII) and aldosterone serum levels. Both lisinopril (ACEi) and MSC treatment prevented monocyte infiltration, reduced tubular cell apoptosis, renal fibrosis and TGFβ expression. Combined therapy provided a further suppression of monocyte infiltration and tubular injury. Lisinopril alone caused a rebound activation of Renin-Angiotensin System (RAS), while MSC suppressed RENmRNA and Renin synthesis and induced a decrease of AII and aldosterone serum levels. Furthermore, in in-vitro and in-vivo experiments, MSC inhibit Human antigen R (HuR) trascription, an enhancer of RENmRNA stability by IL10 release. In conclusion, we demonstrate that in UUO MSC prevent fibrosis, by decreasing HuR-dependent RENmRNA stability. Our findings give a clue to understand the molecular mechanism through which MSC may prevent fibrosis in a wide and heterogeneous number of diseases that share RAS activation as common upstream pathogenic mechanism.
Collapse
Affiliation(s)
- Marilena Gregorini
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Valeria Corradetti
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
- * E-mail:
| | - Chiara Rocca
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Eleonora Francesca Pattonieri
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Teresa Valsania
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Samantha Milanesi
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Nicoletta Serpieri
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Giulia Bedino
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Pasquale Esposito
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Carmelo Libetta
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Maria Antonietta Avanzini
- Laboratory of Transplant Immunology/Cell Factory Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Melissa Mantelli
- Laboratory of Transplant Immunology/Cell Factory Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Daniela Ingo
- Laboratory of Transplant Immunology/Cell Factory Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Sabrina Peressini
- Clinical Chemistry Laboratory Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Riccardo Albertini
- Clinical Chemistry Laboratory Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonio Dal Canton
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Teresa Rampino
- Unit of Nephrology, Dialysis, Transplantation, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| |
Collapse
|
23
|
Shen M, Liu X, Zhang H, Guo SW. Transforming growth factor β1 signaling coincides with epithelial-mesenchymal transition and fibroblast-to-myofibroblast transdifferentiation in the development of adenomyosis in mice. Hum Reprod 2015; 31:355-69. [PMID: 26689216 DOI: 10.1093/humrep/dev314] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 11/25/2015] [Indexed: 12/18/2022] Open
Abstract
STUDY QUESTION Do platelets have any role in the development of adenomyosis? SUMMARY ANSWER Activated platelets coincide with the release of transforming growth factor (TGF)-β1 and induction of the TGF-β/Smad signaling pathway as well as evidence of epithelial-mesenchymal transition (EMT) and fibroblast-to-myofibroblast transdifferentiation (FMT) in a mouse model of adenomyosis, resulting ultimately in fibrosis, as in adenomyosis. WHAT IS KNOWN ALREADY Both EMT and FMT are known to play vital roles in fibrogenesis in general and in endometriosis in particular. EMT has been implicated in the development of adenomyosis, but this was based primarily on cross-sectional observation. It is unclear as to whether adenomyotic lesions and their microenvironment have the machinery to promote EMT and FMT, resulting ultimately in fibrosis. There has not been any published study on the role of platelets in the development of adenomyosis, even though adenomyotic lesions undergo repeated cycles of tissue injury and repair, which implicates the involvement of platelets and constitutes an environment conducive for fibrogenesis. STUDY DESIGN, SIZE, DURATION Adenomyosis was induced in 28 female ICR mice by neonatal dosing of tamoxifen. Another 32 were neonatally dosed without tamoxifen. These mice were sacrificed serially and their tissue samples were subsequently evaluated. PARTICIPANTS/MATERIALS, SETTING, METHODS Female ICR mice with and without induced adenomyosis were sacrificed in batch at 5, 10, 15, 42 and 60 days of age. The depth of myometrial infiltration of endometrial tissues was assessed and immunohistochemistry analysis of biomarkers of EMT and FMT, as well as TGF-β1, phosphorylated Smad3 (p-Smad3) and markers of proliferation, angiogenesis and extracellular matrix (ECM) deposits was performed in ectopic (for adenomyotic mice) and eutopic (controls) endometrial tissue samples. Masson trichrome and Van Gieson stainings were performed to quantify the extent of fibrosis in lesions. Progesterone receptor isoform B (PR-B) staining also was performed. MAIN RESULTS AND THE ROLE OF CHANCE While TGF-β1 immunoreactivity was consistently low in control endometrium, its level was increased dramatically starting from Day 10, along with the extent of platelet aggregation. Staining for TGF-β1 and p-Smad3 increased progressively as adenomyosis progressed, along with markers for proliferation, angiogenesis and ECM deposits. Consistently, staining of vimentin (a marker for stromal or mesenchymal cells) was also increased while that of E-cadherin (a marker for epithelial cells) was reduced. PR-B staining also decreased progressively. Starting from Day 42, α-SMA staining, a marker for myofibroblasts, was elevated in lesions, while in control endometrium, it was negative. Concomitantly, the extent of fibrosis also was increased. LIMITATIONS, REASONS FOR CAUTION This study is limited by the use of histochemistry and immunohistochemistry analyses only and the lack of intervention. WIDER IMPLICATIONS OF THE FINDINGS Like their endometriotic counterpart, adenomyotic lesions and their microenvironment may contain all the necessary molecular machinery to promote fibrogenesis. Platelet-induced activation of the TGF-β/Smad signaling pathway may be a driving force in EMT and FMT in the development of adenomyosis, leading to fibrosis. This study provides the first piece of evidence that adenomyotic lesions are wounds that undergo repeated injury and healing, and as such, platelets play critical roles in the development of adenomyosis. It suggests the potential for the use of anti-platelet therapy in the treatment of adenomyosis, and also opens a new venue for developing novel biomarkers for diagnostic or prognostic purposes. STUDY FUNDING/COMPETING INTERESTS Support for data collection and analysis was provided by grants from the National Science Foundation of China. None of the authors has anything to disclose.
Collapse
Affiliation(s)
- Minhong Shen
- Shanghai Obstetrics/Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Xishi Liu
- Shanghai Obstetrics/Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai 200011, China Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai 200011, China
| | - Hongqi Zhang
- Department of Anatomy, Histology and Embryology, Shanghai Medical Collage, Fudan University, Shanghai 200032, China
| | - Sun-Wei Guo
- Shanghai Obstetrics/Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai 200011, China Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai 200011, China
| |
Collapse
|
24
|
Kim CS, Park JS, Ahn CW, Kim KR. All-Trans Retinoic Acid Has a Potential Therapeutic Role for Diabetic Nephropathy. Yonsei Med J 2015; 56:1597-603. [PMID: 26446642 PMCID: PMC4630048 DOI: 10.3349/ymj.2015.56.6.1597] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 03/03/2015] [Accepted: 04/01/2015] [Indexed: 11/27/2022] Open
Abstract
PURPOSE The aim of this study was to examine the effects of all-trans retinoic acid (ATRA) on diabetic nephropathy. MATERIALS AND METHODS We measured amounts of urinary albumin excretion (UAE) after administrating ATRA to Otsuka Long-Evans Tokushima Fatty (OLETF) rats. In order to understand the mechanism of action for ATRA, we administrated ATRA to examine its inhibitory action on the production of transforming growth factor-β₁ (TGF-β₁), protein kinase C (PKC), and reactive oxidative stress (ROS) in cultured rat mesangial cells (RMCs). RESULTS After 16 weeks of treatment, UAE was lower in the ATRA-treated OLETF rats than in the non-treated OLETF rats (0.07±0.03 mg/mgCr vs. 0.17±0.15 mg/mgCr, p<0.01). After incubation of RMCs in media containing 30 or 5 mM of glucose, treatment with ATRA showed time- and dose-dependent decreases in TGF-β₁ levels and ROS. Moreover, ATRA treatment showed a dose-dependent decrease in PKC expression. CONCLUSION ATRA treatment suppressed UAE and TGF-β₁ synthesis, which was mediated by significant reductions in PKC activity and ROS production. Our results suggest that ATRA has a potential therapeutic role for diabetic nephropathy.
Collapse
Affiliation(s)
- Chul Sik Kim
- Department of Internal Medicine, Hallym University College of Medicine, Anyang, Korea.
| | - Jong Suk Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Chul Woo Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Kyung Rae Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Regulation of MUTYH, a DNA Repair Enzyme, in Renal Proximal Tubular Epithelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:682861. [PMID: 26576226 PMCID: PMC4630661 DOI: 10.1155/2015/682861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/10/2015] [Accepted: 01/17/2015] [Indexed: 12/30/2022]
Abstract
MUTYH is a DNA repair enzyme that initiates a base excision repair (BER) by recognizing and removing 8-Oxoguanine (8-oxoG) and its paired adenine. We demonstrated that both TGF-β1 and H2O2 treatment led to an increased 8-oxoG in cultured human proximal tubule epithelial (HK-2) cells, while the former induced epithelial-mesenchymal transition and the latter caused cell apoptosis. Without stimulation, HK-2 cells showed MUTYH expression in mitochondria. TGF-β1 triggered a transient upregulation of mitochondrial MUTYH and induced the expression of nuclear isoforms, while H2O2 showed no role on MUTYH expression. Ureteral obstruction (UUO) mice exhibited high 8-oxoG reactivity with tubulointerstitial lesions. After obstruction, the MUTYH expression was increased only in tubules at day 3 and decreased with obvious tubular atrophy at day 10. Particularly, MUTYH was primarily located in normal tubular cytoplasm with a dominant mitochondrial form. A few cells with nuclear MUTYH expression were observed in the fibrotic interstitium. We confirmed that increased MUTYH expression was upregulated and positively correlated with the severity of kidney fibrosis. Thus, renal fibrosis caused a cell-type-specific and time-dependent response of oxidative DNA repairs, even within the same tissues. It suggests that intervention of MUTYH might be effective for therapies.
Collapse
|
26
|
Cavaglieri RC, Day RT, Feliers D, Abboud HE. Metformin prevents renal interstitial fibrosis in mice with unilateral ureteral obstruction. Mol Cell Endocrinol 2015; 412:116-22. [PMID: 26067231 DOI: 10.1016/j.mce.2015.06.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/04/2015] [Accepted: 06/04/2015] [Indexed: 01/08/2023]
Abstract
Unilateral ureteral obstruction causes important tubulo-interstitial fibrosis in the kidney. Metformin reduces fibrosis in mice with diabetic nephropathy. We examined the effects of metformin in a mouse model of unilateral ureteral obstruction (UUO). Expression of inflammation and fibrosis markers was studied by immunohistochemistry, immunoblot and quantitative real-time polymerase chain reaction. Seven days after UUO, kidneys presented dilated tubules, expansion of the tubulo-interstitial compartment, and significant infiltration of inflammatory cells. Macrophage infiltration and inflammation markers expression were increased in obstructed kidneys and reduced by metformin. Metformin reduced expression of extracellular matrix proteins and profibrotic factor TGFβ in obstructed kidneys, measured by immunohistochemistry. Interstitial fibroblast activation was evident in obstructed kidneys and ameliorated by metformin. UUO did not affect adenosine monophosphate-activated kinase (AMPK) activity, but metformin activated AMPK. Our results show that metformin prevents or slows down the onset of renal inflammation and fibrosis in mice with UUO, an effect that could be mediated by activation of AMPK.
Collapse
Affiliation(s)
- Rita C Cavaglieri
- Department of Medicine/Nephrology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Robert T Day
- Department of Medicine/Nephrology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Denis Feliers
- Department of Medicine/Nephrology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA.
| | - Hanna E Abboud
- Department of Medicine/Nephrology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
27
|
Mutsaers HAM, Stribos EGD, Glorieux G, Vanholder R, Olinga P. Chronic Kidney Disease and Fibrosis: The Role of Uremic Retention Solutes. Front Med (Lausanne) 2015; 2:60. [PMID: 26380262 PMCID: PMC4553389 DOI: 10.3389/fmed.2015.00060] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/17/2015] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease (CKD) is a major global health concern, and the uremic state is highly associated with fibrogenesis in several organs and tissues. Fibrosis is characterized by excessive production and deposition of extracellular matrix proteins with a detrimental impact on organ function. Another key feature of CKD is the retention and subsequent accumulation of solutes that are normally cleared by the healthy kidney. Several of these uremic retention solutes, including indoxyl sulfate and p-cresyl sulfate, have been suggested to be CKD-specific triggers for the development and perpetuation of fibrosis. The purpose of this brief review is to gather and discuss the current body of evidence linking uremic retention solutes to the fibrotic response during CKD, with a special emphasis on the pathophysiological mechanisms in the kidney.
Collapse
Affiliation(s)
- Henricus A M Mutsaers
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , Netherlands
| | - Elisabeth G D Stribos
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , Netherlands ; Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen , Groningen , Netherlands
| | - Griet Glorieux
- Renal Division, Department of Internal Medicine, Ghent University Hospital , Ghent , Belgium
| | - Raymond Vanholder
- Renal Division, Department of Internal Medicine, Ghent University Hospital , Ghent , Belgium
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , Netherlands
| |
Collapse
|
28
|
Burke M, Pabbidi MR, Farley J, Roman RJ. Molecular mechanisms of renal blood flow autoregulation. Curr Vasc Pharmacol 2015; 12:845-58. [PMID: 24066938 PMCID: PMC4416696 DOI: 10.2174/15701611113116660149] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 12/18/2011] [Accepted: 07/02/2013] [Indexed: 01/10/2023]
Abstract
Diabetes and hypertension are the leading causes of chronic kidney disease and their incidence is increasing at
an alarming rate. Both are associated with impairments in the autoregulation of renal blood flow (RBF) and greater transmission
of fluctuations in arterial pressure to the glomerular capillaries. The ability of the kidney to maintain relatively
constant blood flow, glomerular filtration rate (GFR) and glomerular capillary pressure is mediated by the myogenic response
of afferent arterioles working in concert with tubuloglomerular feedback that adjusts the tone of the afferent arteriole
in response to changes in the delivery of sodium chloride to the macula densa. Despite intensive investigation, the factors
initiating the myogenic response and the signaling pathways involved in the myogenic response and tubuloglomerular
feedback remain uncertain. This review focuses on current thought regarding the molecular mechanisms underlying myogenic
control of renal vascular tone, the interrelationships between the myogenic response and tubuloglomerular feedback,
the evidence that alterations in autoregulation of RBF contributes to hypertension and diabetes-induced nephropathy and
the identification of vascular therapeutic targets for improved renoprotection in hypertensive and diabetic patients.
Collapse
Affiliation(s)
| | | | | | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA.
| |
Collapse
|
29
|
Ai J, Nie J, He J, Guo Q, Li M, Lei Y, Liu Y, Zhou Z, Zhu F, Liang M, Cheng Y, Hou FF. GQ5 Hinders Renal Fibrosis in Obstructive Nephropathy by Selectively Inhibiting TGF-β-Induced Smad3 Phosphorylation. J Am Soc Nephrol 2015; 26:1827-38. [PMID: 25392233 PMCID: PMC4520163 DOI: 10.1681/asn.2014040363] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 09/29/2014] [Indexed: 11/03/2022] Open
Abstract
TGF-β1, via Smad-dependent or Smad-independent signaling, has a central role in the pathogenesis of renal fibrosis. This pathway has been recognized as a potential target for antifibrotic therapy. Here, we identified GQ5, a small molecular phenolic compound isolated from the dried resin of Toxicodendron vernicifluum, as a potent and selective inhibitor of TGF-β1-induced Smad3 phosphorylation. In TGF-β1-stimulated renal tubular epithelial cells and interstitial fibroblast cells, GQ5 inhibited the interaction of Smad3 with TGF-β type I receptor (TβRI) by blocking binding of Smad3 to SARA, suppressed subsequent phosphorylation of Smad3, reduced nuclear translocation of Smad2, Smad3, and Smad4, and downregulated the transcription of major fibrotic genes such as α-smooth muscle actin (α-SMA), collagen I, and fibronectin. Notably, intraperitoneal administration of GQ5 in rats immediately after unilateral ureteral obstruction (UUO) selectively inhibited Smad3 phosphorylation in UUO kidneys, suppressed renal expression of α-SMA, collagen I, and fibronectin, and resulted in impressive renal protection after obstructive injury. Late administration of GQ5 also effectively attenuated fibrotic lesions in obstructive nephropathy. In conclusion, our results suggest that GQ5 hinders renal fibrosis in rats by selective inhibition of TGF-β1-induced Smad3 phosphorylation.
Collapse
Affiliation(s)
- Jun Ai
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Jing Nie
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Jiangbo He
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Qin Guo
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Mei Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Ying Lei
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Zhanmei Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Fengxin Zhu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Min Liang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| | - Yongxian Cheng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, and Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; and
| |
Collapse
|
30
|
Atkinson JM, Pullen N, Da Silva-Lodge M, Williams L, Johnson TS. Inhibition of Thrombin-Activated Fibrinolysis Inhibitor Increases Survival in Experimental Kidney Fibrosis. J Am Soc Nephrol 2015; 26:1925-37. [PMID: 25411467 PMCID: PMC4520161 DOI: 10.1681/asn.2014030303] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 09/30/2014] [Indexed: 11/03/2022] Open
Abstract
Uncontrolled diabetes, inflammation, and hypertension are key contributors to progressive renal fibrosis and subsequent loss of renal function. Reduced fibrinolysis appears to be a feature of ESRD, but its contribution to the fibrotic program has not been extensively studied. Here, we show that in patients with CKD, the activity levels of serum thrombin-activated fibrinolysis inhibitor and plasmin strongly correlated with the degree of renal function impairment. We made similar observations in rats after subtotal nephrectomy and tested whether pharmacologic inhibition of thrombin-activated fibrinolysis inhibitor with UK-396082 could reduce renal fibrosis and improve renal function. Compared with untreated animals, UK-396082-treated animals had reduced glomerular and tubulointerstitial fibrosis after subtotal nephrectomy. Renal function, as measured by an increase in creatinine clearance, was maintained and the rate of increase in proteinuria was reduced in UK-396082-treated animals. Furthermore, cumulative survival improved from 16% to 80% with inhibition of thrombin-activated fibrinolysis inhibitor. Taken together, these data support the importance of the fibrinolytic axis in regulating renal fibrosis and point to a potentially important therapeutic role for suppression of thrombin-activated fibrinolysis inhibitor activity.
Collapse
Affiliation(s)
- John M Atkinson
- Sheffield Kidney Institute & Academic Nephrology Unit, University of Sheffield, Sheffield, United Kingdom; UCB Celltech Pharmaceuticals, Berkshire, United Kingdom; and
| | - Nick Pullen
- Pfizer Global Research Development, Cambridge, Massachusetts
| | - Michelle Da Silva-Lodge
- Sheffield Kidney Institute & Academic Nephrology Unit, University of Sheffield, Sheffield, United Kingdom
| | - Lynne Williams
- Sheffield Kidney Institute & Academic Nephrology Unit, University of Sheffield, Sheffield, United Kingdom
| | - Tim S Johnson
- Sheffield Kidney Institute & Academic Nephrology Unit, University of Sheffield, Sheffield, United Kingdom; UCB Celltech Pharmaceuticals, Berkshire, United Kingdom; and
| |
Collapse
|
31
|
Abstract
Tubulointerstitial fibrosis and glomerulosclerosis, are a major feature of end stage chronic kidney disease (CKD), characterised by an excessive accumulation of extracellular matrix (ECM) proteins. Transforming growth factor beta-1 (TGF-β1) is a cytokine with an important role in many steps of renal fibrosis such as myofibroblast activation and proliferation, ECM protein synthesis and inflammatory cell infiltration. Endoglin is a TGF-β co-receptor that modulates TGF-β responses in different cell types. In numerous cells types, such as mesangial cells or myoblasts, endoglin regulates negatively TGF-β-induced ECM protein expression. However, recently it has been demonstrated that 'in vivo' endoglin promotes fibrotic responses. Furthermore, several studies have demonstrated an increase of endoglin expression in experimental models of renal fibrosis in the kidney and other tissues. Nevertheless, the role of endoglin in renal fibrosis development is unclear and a question arises: Does endoglin protect against renal fibrosis or promotes its development? The purpose of this review is to critically analyse the recent knowledge relating to endoglin and renal fibrosis. Knowledge of endoglin role in this pathology is necessary to consider endoglin as a possible therapeutic target against renal fibrosis.
Collapse
|
32
|
Kanno Y, Kawashita E, Kokado A, Kuretake H, Ikeda K, Okada K, Seishima M, Ueshima S, Matsuo O, Matsuno H. α2AP mediated myofibroblast formation and the development of renal fibrosis in unilateral ureteral obstruction. Sci Rep 2014; 4:5967. [PMID: 25095732 PMCID: PMC5380014 DOI: 10.1038/srep05967] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 07/15/2014] [Indexed: 12/16/2022] Open
Abstract
Renal fibrosis is the final common pathway of a wide variety of chronic kidney diseases. Myofibroblast formation via the differentiation of from tissue-resident fibroblasts and bone marrow-derived mesenchymal stem cells (MSCs), and epithelial-to-mesenchymal transition (EMT) is known to play a pivotal role in the development of renal fibrosis. However, the detailed mechanisms underlying this disorder remain unclear. We herein investigated the role of alpha 2-antiplasmin (α2AP) in myofibroblast formation and the development of renal fibrosis. We observed the development of renal fibrosis using unilateral ureteral obstruction (UUO). α2AP had accumulated in the UUO-induced obstructed kidneys and α2AP deficiency attenuated UUO-induced renal fibrosis in mice. The degree of myofibroblast formation in the obstructed kidneys of α2AP(-/-) mice was less than that in α2AP(+/+) mice. In vitro, α2AP induced myofibroblast formation in renal tubular epithelial cells (RTECs), renal fibrosblasts, and bone marrow-derived mesenchymal stem cells (MSCs). α2AP also induced the production of TGF-β, which is known to be a key regulator of myofibroblast formation and fibrosis. α2AP-induced the TGF-β production was significantly reduced by SP600125, c-Jun N-terminal kinase (JNK) specific inhibitor. Our findings suggest that α2AP induces myofibroblast formation in the obstructed kidneys, and mediates the development of renal fibrosis.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's Collage of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto, 610-0395, Japan
| | - Eri Kawashita
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's Collage of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto, 610-0395, Japan
| | - Akiko Kokado
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's Collage of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto, 610-0395, Japan
| | - Hiromi Kuretake
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's Collage of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto, 610-0395, Japan
| | - Kanako Ikeda
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's Collage of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto, 610-0395, Japan
| | - Kiyotaka Okada
- Department of Physiology II. Kinki University School of Medicine, Osaka-sayama, Japan
| | - Mariko Seishima
- Department of Dermatology, Gifu University Graduate School of Medicine, Yanagido Gifu, Japan
| | - Shigeru Ueshima
- 1] Department of Physiology II. Kinki University School of Medicine, Osaka-sayama, Japan [2] Department of Food Science and Nutrition, Kinki University School of Agriculture, Nara, Japan
| | - Osamu Matsuo
- Department of Physiology II. Kinki University School of Medicine, Osaka-sayama, Japan
| | - Hiroyuki Matsuno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's Collage of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto, 610-0395, Japan
| |
Collapse
|
33
|
Ikeda Y, Ozono I, Tajima S, Imao M, Horinouchi Y, Izawa-Ishizawa Y, Kihira Y, Miyamoto L, Ishizawa K, Tsuchiya K, Tamaki T. Iron chelation by deferoxamine prevents renal interstitial fibrosis in mice with unilateral ureteral obstruction. PLoS One 2014; 9:e89355. [PMID: 24586712 PMCID: PMC3929716 DOI: 10.1371/journal.pone.0089355] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 01/20/2014] [Indexed: 01/19/2023] Open
Abstract
Renal fibrosis plays an important role in the onset and progression of chronic kidney diseases (CKD). Although several mechanisms underlying renal fibrosis and candidate drugs for its treatment have been identified, the effect of iron chelator on renal fibrosis remains unclear. In the present study, we examined the effect of an iron chelator, deferoxamine (DFO), on renal fibrosis in mice with surgically induced unilateral ureter obstruction (UUO). Mice were divided into 4 groups: UUO with vehicle, UUO with DFO, sham with vehicle, and sham with DFO. One week after surgery, augmented renal tubulointerstitial fibrosis and the expression of collagen I, III, and IV increased in mice with UUO; these changes were suppressed by DFO treatment. Similarly, UUO-induced macrophage infiltration of renal interstitial tubules was reduced in UUO mice treated with DFO. UUO-induced expression of inflammatory cytokines and extracellular matrix proteins was abrogated by DFO treatment. DFO inhibited the activation of the transforming growth factor-β1 (TGF-β1)-Smad3 pathway in UUO mice. UUO-induced NADPH oxidase activity and p22phox expression were attenuated by DFO. In the kidneys of UUO mice, divalent metal transporter 1, ferroportin, and ferritin expression was higher and transferrin receptor expression was lower than in sham-operated mice. Increased renal iron content was observed in UUO mice, which was reduced by DFO treatment. These results suggest that iron reduction by DFO prevents renal tubulointerstitial fibrosis by regulating TGF-β-Smad signaling, oxidative stress, and inflammatory responses.
Collapse
Affiliation(s)
- Yasumasa Ikeda
- Department of Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
- * E-mail:
| | - Iori Ozono
- Department of Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
- Student Lab, The University of Tokushima Faculty of Medicine, Tokushima, Japan
| | - Soichiro Tajima
- Department of Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Mizuki Imao
- Department of Medical Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yuya Horinouchi
- Department of Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yuki Izawa-Ishizawa
- Department of Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yoshitaka Kihira
- Department of Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Licht Miyamoto
- Department of Medical Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Medical Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Toshiaki Tamaki
- Department of Pharmacology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| |
Collapse
|
34
|
Effect of stem cells on renal recovery in rat model of partial unilateral upper ureteric obstruction. Pediatr Surg Int 2014; 30:233-8. [PMID: 24370792 DOI: 10.1007/s00383-013-3456-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Untreated obstructive uropathy produces irreversible renal damage and is an important cause of pediatric renal insufficiency. This study was designed to evaluate the effects of stem cell injection on morphological and pathological changes in the rat kidneys with partial unilateral upper ureteric obstruction (PUUUO). METHODS Wistar rats (n = 30) were operated upon to create a PUUUO by the psoas hitch method and were randomized into Group I (control, n = 15) and Group II (stem cell, n = 15); at day 5, 10 and 15, a subgroup of rats (n = 5) from each group was killed and the kidneys harvested. Pathological and morphological changes in the harvested kidneys were studied and compared between the two groups. RESULTS Morphologically, at day 15, Group II had significantly (p = 0.04) greater cortical thickness (0.48 ± 0.17 vs. 0.38 ± 0.09 mm). Histologically, at day 5, Group II had significantly (p = 0.032) lower peri-pelvic fibrosis. Group II group showed greater peri-pelvic inflammation as compared to Group I (p = 0.05). At day 10, lower grades of peri-pelvic fibrosis (p = 0.08), interstitial fibrosis (p = 0.037) and tubular atrophy (p = 0.05) were seen in the Group II. At day 15, Group II demonstrated significantly lower parenchymal loss (p = 0.037), glomerulosclerosis (p = 0.08), interstitial fibrosis (p = 0.08), tubular atrophy (p = 0.08) and peri-pelvic fibrosis (p = 0.08). CONCLUSIONS In a rat model of PUUUO, stem cell injection prevented detrimental changes in renal pathology and preserved renal parenchymal mass.
Collapse
|
35
|
Li J, Yu J, Liu Y, Hu L, Yang B, Zhou X, Wang R, Liang Y. Expression of the Matrix Metalloproteinases and the Tissue Inhibitor of Metalloproteinase Factors are Affected by Tetramethylpyrazine Treatment in a Renal Interstitial Fibrosis Rat Model. J HARD TISSUE BIOL 2014. [DOI: 10.2485/jhtb.23.309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
36
|
Ma SK, Joo SY, Kim CS, Choi JS, Bae EH, Lee J, Kim SW. Increased Phosphorylation of PI3K/Akt/mTOR in the Obstructed Kidney of Rats with Unilateral Ureteral Obstruction. Chonnam Med J 2013; 49:108-12. [PMID: 24400212 PMCID: PMC3881205 DOI: 10.4068/cmj.2013.49.3.108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 11/04/2013] [Accepted: 11/05/2013] [Indexed: 12/24/2022] Open
Abstract
The present study aimed to investigate changes in the mammalian target of rapamycin (mTOR) signaling pathway in the obstructed kidney of rats with unilateral ureteral obstruction (UUO). Male Sprague-Dawley rats were unilaterally obstructed by ligation of the left proximal ureter for 7 days. Control rats were treated in the same way except that no ligature was made. The expression levels of phosphorylated phosphatidylinositol 3-kinase (PI3K), Akt, and mTOR were determined in the kidney by semiquantitative immunoblotting. The protein expression levels of transforming growth factor (TGF)-β1, Bax, and Bcl-2 were also determined in the kidney. The phosphorylation of PI3K, Akt, and mTOR was increased in the kidney of ureteral obstruction rats compared with the control. In the obstructed kidney, the protein expression of TGF-β1 and Bax was also increased, whereas Bcl-2 expression was decreased. In conclusion, the phosphorylation of PI3K/Akt/mTOR was increased in the obstructed kidney of rats with UUO.
Collapse
Affiliation(s)
- Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Soo Yeon Joo
- Department of Physiology, Chonnam National University Medical School, Gwangju, Korea
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Joon Seok Choi
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Jongun Lee
- Department of Physiology, Chonnam National University Medical School, Gwangju, Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
37
|
Grabias BM, Konstantopoulos K. The physical basis of renal fibrosis: effects of altered hydrodynamic forces on kidney homeostasis. Am J Physiol Renal Physiol 2013; 306:F473-85. [PMID: 24352503 DOI: 10.1152/ajprenal.00503.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Healthy kidneys are continuously exposed to an array of physical forces as they filter the blood: shear stress along the inner lumen of the tubules, distension of the tubular walls in response to changing fluid pressures, and bending moments along both the cilia and microvilli of individual epithelial cells that comprise the tubules. Dysregulation of kidney homeostasis via underlying medical conditions such as hypertension, diabetes, or glomerulonephritis fundamentally elevates the magnitudes of each principle force in the kidney and leads to fibrotic scarring and eventual loss of organ function. The purpose of this review is to summarize the progress made characterizing the response of kidney cells to pathological levels of mechanical stimuli. In particular, we examine important, mechanically responsive signaling cascades and explore fundamental changes in renal cell homeostasis after cyclic strain or fluid shear stress exposure. Elucidating the effects of these disease-related mechanical imbalances on endogenous signaling events in kidney cells presents a unique opportunity to better understand the fibrotic process.
Collapse
Affiliation(s)
- Bryan M Grabias
- Dept. of Chemical and Biomolecular Engineering, The Johns Hopkins Univ., New Engineering Bldg. 114, 3400 N. Charles St., Baltimore, MD 21218.
| | | |
Collapse
|
38
|
Canonical transforming growth factor-β signaling regulates disintegrin metalloprotease expression in experimental renal fibrosis via miR-29. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1885-1896. [PMID: 24103556 DOI: 10.1016/j.ajpath.2013.08.027] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 07/29/2013] [Accepted: 08/21/2013] [Indexed: 01/06/2023]
Abstract
Fibrosis pathophysiology is critically regulated by Smad 2- and Smad 3-mediated transforming growth factor-β (TGF-β) signaling. Disintegrin metalloproteases (Adam) can manipulate the signaling environment, however, the role and regulation of ADAMs in renal fibrosis remain unclear. TGF-β stimulation of renal cells results in a significant up-regulation of Adams 10, 17, 12, and 19. The selective Smad2/3 inhibitor SB 525334 reversed these TGF-β-induced changes. In vivo, using ureteral obstruction to model renal fibrosis, we observed increased Adams gene expression that was blocked by oral administration of SB 525334. Similar increases in Adam gene expression also occurred in preclinical models of hypertension-induced renal damage and glomerulonephritis. miRNAs are a recently discovered second level of regulation of gene expression. Analysis of 3' untranslated regions of Adam12 and Adam19 mRNAs showed multiple binding sites for miR-29a, miR-29b, and miR-29c. We show that miR-29 family expression is decreased after unilateral ureter obstruction and this significant decrease in miR-29 family expression was observed consistently in preclinical models of renal dysfunction and correlated with an increase in Adam12 and Adam19 expression. Exogenous overexpression of the miR-29 family blocked TGF-β-mediated up-regulation of Adam12 and Adam19 gene expression. This study shows that Adams are involved in renal fibrosis and are regulated by canonical TGF-β signaling and miR-29. Therefore, both Adams and the miR-29 family represent therapeutic targets for renal fibrosis.
Collapse
|
39
|
Kim J, Padanilam BJ. Renal nerves drive interstitial fibrogenesis in obstructive nephropathy. J Am Soc Nephrol 2012; 24:229-42. [PMID: 23264683 DOI: 10.1681/asn.2012070678] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The signals that drive fibrogenesis after an initiating insult to the kidney are incompletely understood. Here, we report that renal nerve stimulation after ureteral obstruction is the primary profibrotic signal and that renal denervation prevents both fibrogenesis and the inflammatory cascade. Local infusion of neural factors, norepinephrine, and calcitonin gene-related peptide (CGRP) in denervated kidneys mimicked the fibrotic response observed in innervated obstructed kidneys. Norepinephrine and CGRP act through the α(2)-adrenergic receptor and CGRP receptor, respectively, because blocking these receptors prevented fibrosis, the inflammatory response, and tubular cell death. In tubular epithelial cells, both norepinephrine and CGRP induced apoptosis and the release of profibrotic factors capable of stimulating the differentiation of fibroblasts to myofibroblasts. In conclusion, these data suggest that nerve-derived signaling molecules may drive renal fibrosis and that their suppression may be a therapeutic approach to fibrosis prevention.
Collapse
Affiliation(s)
- Jinu Kim
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska 68198-5850, USA
| | | |
Collapse
|
40
|
Nlandu Khodo S, Dizin E, Sossauer G, Szanto I, Martin PY, Feraille E, Krause KH, de Seigneux S. NADPH-oxidase 4 protects against kidney fibrosis during chronic renal injury. J Am Soc Nephrol 2012; 23:1967-76. [PMID: 23100220 DOI: 10.1681/asn.2012040373] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
NADPH oxidases synthesize reactive oxygen species that may participate in fibrosis progression. NOX4 and NOX2 are NADPH oxidases expressed in the kidneys, with the former being the major renal isoform, but their contribution to renal disease is not well understood. Here, we used the unilateral urinary obstruction model of chronic renal injury to decipher the role of these enzymes using wild-type, NOX4-, NOX2-, and NOX4/NOX2-deficient mice. Compared with wild-type mice, NOX4-deficient mice exhibited more interstitial fibrosis and tubular apoptosis after obstruction, with lower interstitial capillary density and reduced expression of hypoxia-inducible factor-1α and vascular endothelial growth factor in obstructed kidneys. Furthermore, NOX4-deficient kidneys exhibited increased oxidative stress. With NOX4 deficiency, renal expression of other NOX isoforms was not altered but NRF2 protein expression was reduced under both basal and obstructed conditions. Concomitant deficiency of NOX2 did not modify the phenotype exhibited by NOX4-deficient mice after obstruction. NOX4 silencing in a mouse collecting duct (mCCD(cl1)) cell line increased TGF-β1-induced apoptosis and decreased NRF2 protein along with expression of its target genes. In addition, NOX4 silencing decreased hypoxia-inducible factor-1α and expression of its target genes in response to hypoxia. In summary, these results demonstrate that the absence of NOX4 promotes kidney fibrosis, independent of NOX2, through enhanced tubular cell apoptosis, decreased microvascularization, and enhanced oxidative stress. Thus, NOX4 is crucial for the survival of kidney tubular cells under injurious conditions.
Collapse
Affiliation(s)
- Stellor Nlandu Khodo
- Department of Cell Physiology and Metabolism, University Medical Center, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Renal IL-18 production is macrophage independent during obstructive injury. PLoS One 2012; 7:e47417. [PMID: 23077611 PMCID: PMC3470595 DOI: 10.1371/journal.pone.0047417] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 09/14/2012] [Indexed: 12/11/2022] Open
Abstract
Background Interleukin 18 (IL-18) is a pro-inflammatory cytokine that mediates fibrotic renal injury during obstruction. Macrophages are a well-known source of IL-18; however, renal tubular epithelial cells are also a potential source of this cytokine. We hypothesized that IL-18 is predominantly a renal tubular cell product and is produced during renal obstruction independent of macrophage infiltration. Methods To study this, male C57BL6 mice were subjected to unilateral ureteral obstruction (UUO) vs. sham operation in the presence or absence of macrophage depletion (liposomal clodronate (1 ml/100 g body weight i.v.)). The animals were sacrificed 1 week after surgery and renal cortical tissue harvested. Tissue levels of active IL-18 (ELISA), IL-18 receptor mRNA expression (real time PCR), and active caspase-1 expression (western blot) were measured. The cellular localization of IL-18 and IL-18R was assessed using dual labeling immunofluorescent staining (IFS). Results Immunohistochemical staining of renal tissue sections confirmed macrophage depletion by liposomal clodronate. IL-18 production, IL-18R expression, and active caspase 1 expression were elevated in response to renal obstruction and did not decline to a significant degree in the presence of macrophage depletion. Obstruction-induced IL-18 and IL-18R production localized predominantly to tubular epithelial cells (TEC) during obstruction despite macrophage depletion. Conclusion These results demonstrate that renal tubular epithelial cells are the primary source of IL-18 production during obstructive injury, and that tubular cell production of IL-18 occurs independent of macrophage infiltration.
Collapse
|
42
|
Ito H, Yan X, Nagata N, Aritake K, Katsumata Y, Matsuhashi T, Nakamura M, Hirai H, Urade Y, Asano K, Kubo M, Utsunomiya Y, Hosoya T, Fukuda K, Sano M. PGD2-CRTH2 pathway promotes tubulointerstitial fibrosis. J Am Soc Nephrol 2012; 23:1797-809. [PMID: 22997255 DOI: 10.1681/asn.2012020126] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Urinary excretion of lipocalin-type PGD(2) synthase (L-PGDS), which converts PG H(2) to PGD(2), increases in early diabetic nephropathy. In addition, L-PGDS expression in the tubular epithelium increases in adriamycin-induced nephropathy, suggesting that locally produced L-PGDS may promote the development of CKD. In this study, we found that L-PGDS-derived PGD(2) contributes to the progression of renal fibrosis via CRTH2-mediated activation of Th2 lymphocytes. In a mouse model, the tubular epithelium synthesized L-PGDS de novo after unilateral ureteral obstruction (UUO). L-PGDS-knockout mice and CRTH2-knockout mice both exhibited less renal fibrosis, reduced infiltration of Th2 lymphocytes into the cortex, and decreased production of the Th2 cytokines IL-4 and IL-13. Furthermore, oral administration of a CRTH2 antagonist, beginning 3 days after UUO, suppressed the progression of renal fibrosis. Ablation of IL-4 and IL-13 also ameliorated renal fibrosis in the UUO kidney. Taken together, these data suggest that blocking the activation of CRTH2 by PGD(2) might be a strategy to slow the progression of renal fibrosis in CKD.
Collapse
Affiliation(s)
- Hideyuki Ito
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Transforming growth factor-β induces vascular endothelial growth factor-C expression leading to lymphangiogenesis in rat unilateral ureteral obstruction. Kidney Int 2012; 81:865-79. [PMID: 22258325 DOI: 10.1038/ki.2011.464] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammation is recognized as an important contributor to lymphangiogenesis; however, in tubulointerstitial lesions in human chronic kidney diseases, this process is better correlated with the presence of myofibroblasts rather than macrophages. As little is known about the interaction between lymphangiogenesis and renal fibrosis, we utilized the rat unilateral ureteral obstruction model to analyze inflammation, fibrosis, lymphangiogenesis, and growth factor expression. Additionally, we determined the relationship between vascular endothelial growth factor-C (VEGF-C), an inducer of lymphangiogenesis, and the profibrotic factor, transforming growth factor-β1 (TGF-β1). The expression of both TGF-β1 and VEGF-C was detected in tubular epithelial and mononuclear cells, and gradually increased, peaking 14 days after ureteral obstruction. The kinetics and localization of VEGF-C were similar to those of TGF-β1, and the expression of these growth factors and lymphangiogenesis were linked with the progression of fibrosis. VEGF-C expression was upregulated by TGF-β1 in cultured proximal tubular epithelial cells, collecting duct cells, and macrophages. Both in vitro and in vivo, the induction of VEGF-C along with the overall appearance of lymphatics in vivo was specifically suppressed by the TGF-β type I receptor inhibitor LY364947. Thus, TGF-β1 induces VEGF-C expression, which leads to lymphangiogenesis.
Collapse
|
45
|
Samarakoon R, Overstreet JM, Higgins SP, Higgins PJ. TGF-β1 → SMAD/p53/USF2 → PAI-1 transcriptional axis in ureteral obstruction-induced renal fibrosis. Cell Tissue Res 2012; 347:117-28. [PMID: 21638209 PMCID: PMC3188682 DOI: 10.1007/s00441-011-1181-y] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 04/15/2011] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease constitutes an increasing medical burden affecting 26 million people in the United States alone. Diabetes, hypertension, ischemia, acute injury, and urological obstruction contribute to renal fibrosis, a common pathological hallmark of chronic kidney disease. Regardless of etiology, elevated TGF-β1 levels are causatively linked to the activation of profibrotic signaling pathways initiated by angiotensin, glucose, and oxidative stress. Unilateral ureteral obstruction (UUO) is a useful and accessible model to identify mechanisms underlying the progression of renal fibrosis. Plasminogen activator inhibitor-1 (PAI-1), a major effector and downstream target of TGF-β1 in the progression of several clinically important fibrotic disorders, is highly up-regulated in UUO and causatively linked to disease severity. SMAD and non-SMAD pathways (pp60(c-src), epidermal growth factor receptor [EGFR], mitogen-activated protein kinase, p53) are required for PAI-1 induction by TGF-β1. SMAD2/3, pp60(c-src), EGFR, and p53 activation are each increased in the obstructed kidney. This review summarizes the molecular basis and translational significance of TGF-β1-stimulated PAI-1 expression in the progression of kidney disease induced by ureteral obstruction. Mechanisms discussed here appear to be operative in other renal fibrotic disorders and are relevant to the global issue of tissue fibrosis, regardless of organ site.
Collapse
Affiliation(s)
- Rohan Samarakoon
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany NY 12208, USA
| | - Jessica M. Overstreet
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany NY 12208, USA
| | - Stephen P. Higgins
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany NY 12208, USA
| | - Paul J. Higgins
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany NY 12208, USA
| |
Collapse
|
46
|
The kinase Pyk2 is involved in renal fibrosis by means of mechanical stretch-induced growth factor expression in renal tubules. Kidney Int 2011; 81:449-57. [PMID: 22157654 DOI: 10.1038/ki.2011.403] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Unilateral ureteral obstruction is a well-established experimental model of progressive renal fibrosis. We tested whether mechanical stretch and subsequent renal tubular distension might lead to renal fibrosis by first studying renal tubular epithelial cells in culture. We found that mechanical stretch induced reactive oxygen species that in turn activated the cytoplasmic proline-rich tyrosine kinase-2 (Pyk2). This kinase is abundantly expressed in tubular epithelial cells where it is activated by several stimuli. Using mice with deletion of Pyk2 we found that the expression of transforming growth factor-β1 induced by mechanical stretch in renal tubular epithelial cells was significantly reduced. The expression of connective tissue growth factor was also reduced in the Pyk2(-/-) mice. We also found that expression of connective tissue growth factor was independent of transforming growth factor-β1, but dependent on the Rho-associated coiled-coil forming protein kinase pathway. Thus, Pyk2 may be an important initiating factor in renal fibrosis and might be a new therapeutic target for ameliorating renal fibrosis.
Collapse
|
47
|
Chiang CK, Sheu ML, Lin YW, Wu CT, Yang CC, Chen MW, Hung KY, Wu KD, Liu SH. Honokiol ameliorates renal fibrosis by inhibiting extracellular matrix and pro-inflammatory factors in vivo and in vitro. Br J Pharmacol 2011; 163:586-97. [PMID: 21265825 DOI: 10.1111/j.1476-5381.2011.01242.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Renal fibrosis acts as the common pathway leading to the development of end-stage renal disease. The present study investigated, in vivo and in vitro, the anti-fibrotic and anti-inflammatory effects, particularly on the epithelial to mesenchymal transition of renal tubular cells, exerted by honokiol, a phytochemical used in traditional medicine, and mechanisms underlying these effects. EXPERIMENTAL APPROACH Anti-fibrotic effects in vivo were assayed in a rat model of renal fibrosis [the unilateral ureteral obstruction (UUO) model]. A rat tubular epithelial cell line (NRK-52E) was stimulated by transforming growth factor-β1 (TGF-β1) and treated with honokiol to explore possible mechanisms of these anti-fibrotic effects. Gene or protein expression was analysed by Northern or Western blotting. Transcriptional regulation was investigated using luciferase activity driven by a connective tissue growth factor (CTGF) promoter. KEY RESULTS Honokiol slowed development of renal fibrosis both in vivo and in vitro. Honokiol treatment attenuated tubulointerstitial fibrosis and expression of pro-fibrotic factors in the UUO model. Honokiol also decreased expression of the mRNA for the chemokine CCL2 and for the intracellular adhesion molecule-1, as well as accumulation of type I (α1) collagen and fibronectin in UUO kidneys. Phosphorylation of Smad-2/3 induced by TGF-β1 and CTGF luciferase activity in renal tubular cells were also inhibited by honokiol. CONCLUSIONS AND IMPLICATIONS Honokiol suppressed expression of pro-fibrotic and pro-inflammatory factors and of extracellular matrix proteins. Honokiol may become a therapeutic agent to prevent renal fibrosis.
Collapse
Affiliation(s)
- Chih-Kang Chiang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
VanderBrink BA, Asanuma H, Hile K, Zhang H, Rink RC, Meldrum KK. Interleukin-18 stimulates a positive feedback loop during renal obstruction via interleukin-18 receptor. J Urol 2011; 186:1502-8. [PMID: 21855933 DOI: 10.1016/j.juro.2011.05.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Indexed: 12/11/2022]
Abstract
PURPOSE Interleukin-18 is a proinflammatory cytokine that is an important mediator of obstruction induced renal tubulointerstitial fibrosis independent of tumor necrosis factor-α and β1 activity. We hypothesized that interleukin-18 stimulates a positive feedback loop during obstruction via interleukin-18 receptor to increase interleukin-18 gene expression and protein production. MATERIALS AND METHODS Male C57BL6 interleukin-18 receptor knockout (The Jackson Laboratory, Bar Harbor, Maine) and control wild-type mice underwent unilateral ureteral obstruction or sham operation and were sacrificed 1 week after surgery. Renal cortical tissue samples were harvested and analyzed for interleukin-18 protein by enzyme-linked immunosorbent assay, and for interleukin-18 and interleukin-18 receptor gene expression by quantitative polymerase chain reaction. The specific cellular localization of interleukin-18 and interleukin-18 receptor expression during obstruction was assessed using dual labeling immunofluorescence staining. RESULTS Renal interleukin-18 receptor expression increased significantly in wild-type mice in response to obstruction but remained at sham operation levels in interleukin-18 receptor knockout mice. Similarly while interleukin-18 protein and gene expression were significantly increased in wild-type mice in response to obstruction, interleukin-18 levels and gene expression were significantly decreased during obstruction in knockout mice. Obstruction induced interleukin-18 and interleukin-18 receptor production were localized predominantly to tubular epithelial cells and to a lesser extent to the renal interstitium. CONCLUSIONS Results reveal that interleukin-18 stimulates a positive feedback loop via interleukin-18 receptor during renal obstruction to stimulate interleukin-18 production and gene expression. The predominant cellular source of interleukin-18 production during renal obstruction appears to be tubular epithelial cells rather than infiltrating macrophages.
Collapse
Affiliation(s)
- Brian A VanderBrink
- Department of Urology, James Whitcomb Riley Hospital for Children, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | |
Collapse
|
49
|
Morishita Y, Watanabe M, Nakazawa E, Ishibashi K, Kusano E. The interaction of LFA-1 on mononuclear cells and ICAM-1 on tubular epithelial cells accelerates TGF-β1-induced renal epithelial-mesenchymal transition. PLoS One 2011; 6:e23267. [PMID: 21850266 PMCID: PMC3151298 DOI: 10.1371/journal.pone.0023267] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 07/14/2011] [Indexed: 12/24/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) of renal epithelial cells (RTECs) has pivotal roles in the development of renal fibrosis. Although the interaction of lymphocyte function-associated antigen 1 (LFA-1) on leukocytes and its ligand, intracellular adhesion molecule 1 (ICAM-1), plays essential roles in most inflammatory reactions, its pathogenetic role in the EMT of RTECs remains to be clarified. In the present study, we investigated the effect of the interaction of LFA-1 on peripheral blood mononuclear cells (PBMCs) and ICAM-1 on HK-2 cells after stimulation with TGF-β(1) on the EMT of RTECs. ICAM-1 was highly expressed in HK-2 cells. After TGF-β(1) stimulation, the chemokines CCL3 and CXCL12 increased on HK-2 cells. After co-culture of PBMCs and HK-2 cells pre-stimulated with TGF-β(1) (0.1 ng/ml) (HK-2-TGF-β(1) (0.1)), the expression of the active form of LFA-1 increased on PBMCs; however, total LFA-1 expression did not change. The expression of the active form of LFA-1 on PBMCs did not increase after co-culture with not CCL3 but CXCL12 knockdown HK-2-TGF-β(1) (0.1). The expression of epithelial cell junction markers (E-cadherin and occludin) further decreased and that of mesenchymal markers (vimentin and fibronectin) further increased in HK-2-TGF-β(1) (0.1) after co-culture with PBMCs for 24 hrs (HK-2-TGF-β(1) (0.1)-PBMCs). The phosphorylation of ERK 1/2 but not smad2 and smad3 increased in HK-2-TGF-β(1) (0.1)-PBMCs. The snail and slug signaling did not increase HK-2-TGF-β(1) (0.1)-PBMCs. Although the migration and invasion of HK-2 cells induced full EMT by a high dose (10.0 ng/ml) and long-term (72-96 hrs) TGF-β(1) stimulation increased, that of HK-2-TGF-β(1) (0.1)-PBMCs did not increase. These results suggested that HK-2 cells stimulated with TGF-β(1) induced conformational activation of LFA-1 on PBMCs by increased CXCL12. Then, the direct interaction of LFA-1 on PBMCs and ICAM-1 on HK-2 cells activated ERK1/2 signaling to accelerate the part of EMT of HK-2 cells induced by TGF-β(1).
Collapse
Affiliation(s)
- Yoshiyuki Morishita
- Division of Nephrology, Department of Medicine, Jichi Medical University, Tochigi, Japan.
| | | | | | | | | |
Collapse
|
50
|
Denby L, Ramdas V, McBride MW, Wang J, Robinson H, McClure J, Crawford W, Lu R, Hillyard DZ, Khanin R, Agami R, Dominiczak AF, Sharpe CC, Baker AH. miR-21 and miR-214 are consistently modulated during renal injury in rodent models. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:661-72. [PMID: 21704009 PMCID: PMC3157202 DOI: 10.1016/j.ajpath.2011.04.021] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 04/01/2011] [Accepted: 04/29/2011] [Indexed: 12/19/2022]
Abstract
Transforming growth factor (TGF)-β is one of the main fibrogenic cytokines that drives the pathophysiology of progressive renal scarring. MicroRNAs (miRNAs) are endogenous non-coding RNAs that post-transcriptionally regulate gene expression. We examined the role of TGF-β-induced expression of miR-21, miRNAs in cell culture models and miRNA expression in relevant models of renal disease. In vitro, TGF-β changed expression of miR-21, miR-214, and miR-145 in rat mesangial cells (CRL-2753) and miR-214, miR-21, miR-30c, miR-200b, and miR-200c during induction of epithelial-mesenchymal transition in rat tubular epithelial cells (NRK52E). miR-214 expression was robustly modulated in both cell types, whereas in tubular epithelial cells miR-21 was increased and miR-200b and miR-200c were decreased by 58% and 48%, respectively, in response to TGF-β. TGF-β receptor-1 was found to be a target of miR-200b/c and was down-regulated after overexpression of miR-200c. To assess the differential expression of these miRNAs in vivo, we used the anti-Thy1.1 mesangial glomerulonephritis model and the unilateral ureteral obstruction model in which TGF-β plays a role and also a genetic model of hypertension, the stroke-prone spontaneously hypertensive rat with and without salt loading. The expressions of miR-214 and miR-21 were significantly increased in all in vivo models, showing a possible miRNA signature of renal damage despite differing causes.
Collapse
Affiliation(s)
- Laura Denby
- BHF Glasgow Cardiovascular Research Centre, Glasgow, United Kingdom
| | - Vasudev Ramdas
- BHF Glasgow Cardiovascular Research Centre, Glasgow, United Kingdom
| | | | - Joe Wang
- Department of Renal Medicine, The Rayne Institute, King's College London, London, United Kingdom
| | - Hollie Robinson
- BHF Glasgow Cardiovascular Research Centre, Glasgow, United Kingdom
| | - John McClure
- BHF Glasgow Cardiovascular Research Centre, Glasgow, United Kingdom
| | - Wendy Crawford
- BHF Glasgow Cardiovascular Research Centre, Glasgow, United Kingdom
| | - Ruifang Lu
- BHF Glasgow Cardiovascular Research Centre, Glasgow, United Kingdom
| | | | - Raya Khanin
- Department of Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Reuven Agami
- Division of Gene Regulation, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Claire C. Sharpe
- Department of Renal Medicine, The Rayne Institute, King's College London, London, United Kingdom
| | - Andrew H. Baker
- BHF Glasgow Cardiovascular Research Centre, Glasgow, United Kingdom
| |
Collapse
|