1
|
Srinivasan S, Sherwood DR. The life cycle of type IV collagen. Matrix Biol 2025:S0945-053X(25)00037-X. [PMID: 40306374 DOI: 10.1016/j.matbio.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Type IV collagen is a large triple helical molecule that forms a covalently cross-linked network within basement membranes (BMs). Type IV collagen networks play key roles in mechanically supporting tissues, shaping organs, filtering blood, and cell signaling. To ensure tissue health and function, all aspects of the type IV collagen life cycle must be carried out accurately. However, the large triple helical structure and complex life-cycle of type IV collagen, poses many challenges to cells and tissues. Type IV collagen predominantly forms heterotrimers and to ensure proper construction, expression of the distinct α-chains that comprise a heterotrimer needs tight regulation. The α-chains must also be accurately modified by several enzymes, some of which are specific to collagens, to build and stabilize the triple helical trimer. In addition, type IV collagen is exceptionally long (400nm) and thus the packaging and trafficking of the triple helical trimer from the ER to the Golgi must be modified to accommodate the large type IV collagen molecule. During ER-to-Golgi trafficking, as well as during secretion and transport in the extracellular space type IV collagen also associates with specific chaperone molecules that maintain the structure and solubility of collagen IV. Type IV collagen trimers are then delivered to BMs from local and distant sources where they are integrated into BMs by interactions with cell surface receptors and many diverse BM resident proteins. Within BMs type IV collagen self-associates into a network and is crosslinked by BM resident enzymes. Finally, homeostatic type IV collagen levels in BMs are maintained by poorly understood mechanisms involving proteolysis and endocytosis. Here, we provide an overview of the life cycle of collagen IV, highlighting unique mechanisms and poorly understood aspects of type IV collagen regulation.
Collapse
Affiliation(s)
- Sandhya Srinivasan
- Department of Biology, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA
| | - David R Sherwood
- Department of Biology, Duke University, 130 Science Drive, Box 90338, Durham, NC 27708, USA.
| |
Collapse
|
2
|
Kukita K, Sakaguchi M, Inoue H, Imamura Y, Shin Y. Type IV collagen expression is regulated by Notch3-mediated Notch signaling during angiogenesis. Biochem Biophys Res Commun 2025; 749:151351. [PMID: 39842335 DOI: 10.1016/j.bbrc.2025.151351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
Angiogenesis, the process of new blood vessel formation, involves endothelial cell proliferation and migration, accompanied by the remodeling of the extracellular matrix (ECM). Type IV collagen, a major ECM component, plays a critical role in vascular basement membrane regeneration, influencing cell polarity, migration, and survival. This study examines the regulatory role of Notch signaling, mediated by Notch3, in type IV collagen expression using TIG-1 fibroblasts and a co-culture angiogenesis model with human umbilical vein endothelial cells (HUVECs). Using small interfering RNA (siRNA) to suppress Notch3 expression, we observed a significant reduction in COL4A1 gene expression, which encodes the α1 chain of type IV collagen. Conversely, transient expression of the Notch3 intracellular domain (NICD3) activated Notch signaling, resulting in increased COL4A1 expression. In the co-culture model, pre-treatment of TIG-1 cells with Notch signaling inhibitors, including siNotch3 and DAPT, decreased the number of α1(IV)-positive TIG-1 fibroblasts adjacent to HUVECs. This reduction highlights the essential role of Notch3-mediated signaling in promoting type IV collagen expression during angiogenesis. Our findings suggest that Notch signaling regulates type IV collagen expression levels, supporting basement membrane formation and vascular maturation. These results provide insight into the molecular mechanisms of angiogenesis, potentially contributing to therapeutic strategies targeting vascular-related pathologies.
Collapse
Affiliation(s)
- Kazuki Kukita
- Graduate School of Engineering, Kogakuin University, Tokyo, Japan
| | - Masayoshi Sakaguchi
- Graduate School of Engineering, Kogakuin University, Tokyo, Japan; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan
| | - Hiroki Inoue
- Department of Medical Sciences, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yasutada Imamura
- Graduate School of Engineering, Kogakuin University, Tokyo, Japan; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan
| | - Yongchol Shin
- Graduate School of Engineering, Kogakuin University, Tokyo, Japan; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, Tokyo, Japan.
| |
Collapse
|
3
|
Bordini M, Mazzoni M, Di Nunzio M, Zappaterra M, Sirri F, Meluzzi A, Petracci M, Soglia F. Time course evaluation of collagen type IV in Pectoralis major muscles of broiler chickens selected for different growth-rates. Poult Sci 2024; 103:103179. [PMID: 37931400 PMCID: PMC10652102 DOI: 10.1016/j.psj.2023.103179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 11/08/2023] Open
Abstract
Collagen type IV (COL4) is one of the major components of animals' and humans' basement membranes of several tissues, such as skeletal muscles and vascular endothelia. Alterations in COL4 assembly and secretion are associated to muscular disorders in humans and animals among which growth-related abnormalities such as white striping and wooden breast affecting Pectoralis major muscles (PMs) in modern fast-growing (FG) chickens. Considering the high prevalence of these myopathies in FG broilers and that a worsening is observed as the bird slaughter age is increased, the present study was intended to evaluate the distribution and the expression level of COL4 protein and its coding genes in PMs of FG broilers at different stages of muscle development (i.e., 7, 14, 21, 28, 35, and 42 d of age). Medium-growing (MG) chickens have been considered as the control group in consideration of the lower selection pressure on breast muscle growth rate and hypertrophy. Briefly, 5 PM/sampling time/genotype were selected for western blot, immunohistochemistry (IHC), and gene expression analyses. The normalized expression levels of COL4 coding genes showed an overexpression of COL4A2 in FG than MG at d 28, as well as a significant decrease in its expression over their rearing period. Overall, results obtained through the gene expression analysis suggested that selection for the hypertrophic growth of FG broilers may have led to an altered regulation of fibroblast proliferation and COL4 synthesis. Moreover, western blot and IHC analyses suggested an altered secretion and/or degradation of COL4 protein in FG broilers, as evidenced by the fluctuating trend of 2 bands observed in FG over time. In view of the above, the present research supports the evidence about a potential aberrant synthesis and/or degradation of COL4 and corroborates the hypothesis regarding a likely involvement of COL4 in the series of events underlying the growth-related abnormalities in modern FG broilers.
Collapse
Affiliation(s)
- Martina Bordini
- Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Maurizio Mazzoni
- Department of Veterinary Medical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Mattia Di Nunzio
- Department of Food, Environmental and Nutritional Sciences (Defens), University of Milan, Milan, 20133, Italy
| | - Martina Zappaterra
- Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Federico Sirri
- Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Adele Meluzzi
- Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Massimiliano Petracci
- Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Francesca Soglia
- Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
4
|
Salzano A, Fioriniello S, D'Onofrio N, Balestrieri ML, Aiese Cigliano R, Neglia G, Della Ragione F, Campanile G. Transcriptomic profiles of the ruminal wall in Italian Mediterranean dairy buffaloes fed green forage. BMC Genomics 2023; 24:133. [PMID: 36941576 PMCID: PMC10029215 DOI: 10.1186/s12864-023-09215-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 02/28/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Green feed diet in ruminants exerts a beneficial effect on rumen metabolism and enhances the content of milk nutraceutical quality. At present, a comprehensive analysis focused on the identification of genes, and therefore, biological processes modulated by the green feed in buffalo rumen has never been reported. We performed RNA-sequencing in the rumen of buffaloes fed a total mixed ration (TMR) + the inclusion of 30% of ryegrass green feed (treated) or TMR (control), and identified differentially expressed genes (DEGs) using EdgeR and NOISeq tools. RESULTS We found 155 DEGs using EdgeR (p-values < 0.05) and 61 DEGs using NOISeq (prob ≥0.8), 30 of which are shared. The rt-qPCR validation suggested a higher reliability of EdgeR results as compared with NOISeq data, in our biological context. Gene Ontology analysis of DEGs identified using EdgeR revealed that green feed modulates biological processes relevant for the rumen physiology and, then, health and well-being of buffaloes, such as lipid metabolism, response to the oxidative stress, immune response, and muscle structure and function. Accordingly, we found: (i) up-regulation of HSD17B13, LOC102410803 (or PSAT1) and HYKK, and down-regulation of CDO1, SELENBP1 and PEMT, encoding factors involved in energy, lipid and amino acid metabolism; (ii) enhanced expression of SIM2 and TRIM14, whose products are implicated in the immune response and defense against infections, and reduced expression of LOC112585166 (or SAAL1), ROR2, SMOC2, and S100A11, encoding pro-inflammatory factors; (iii) up-regulation of NUDT18, DNAJA4 and HSF4, whose products counteract stressful conditions, and down-regulation of LOC102396388 (or UGT1A9) and LOC102413340 (or MRP4/ABCC4), encoding detoxifying factors; (iv) increased expression of KCNK10, CACNG4, and ATP2B4, encoding proteins modulating Ca2+ homeostasis, and reduced expression of the cytoskeleton-related MYH11 and DES. CONCLUSION Although statistically unpowered, this study suggests that green feed modulates the expression of genes involved in biological processes relevant for rumen functionality and physiology, and thus, for welfare and quality production in Italian Mediterranean dairy buffaloes. These findings, that need to be further confirmed through the validation of additional DEGs, allow to speculate a role of green feed in the production of nutraceutical molecules, whose levels might be enhanced also in milk.
Collapse
Affiliation(s)
- Angela Salzano
- Department of Veterinary Medicine and Animal Production, Federico II University, Naples, Italy
| | | | - Nunzia D'Onofrio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | | | - Gianluca Neglia
- Department of Veterinary Medicine and Animal Production, Federico II University, Naples, Italy
| | - Floriana Della Ragione
- Institute of Genetics and Biophysics 'A. Buzzati-Traverso', CNR, Naples, Italy.
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Isernia, Italy.
| | - Giuseppe Campanile
- Department of Veterinary Medicine and Animal Production, Federico II University, Naples, Italy
| |
Collapse
|
5
|
Ikeda SI, Kurihara T, Jiang X, Miwa Y, Lee D, Serizawa N, Jeong H, Mori K, Katada Y, Kunimi H, Ozawa N, Shoda C, Ibuki M, Negishi K, Torii H, Tsubota K. Scleral PERK and ATF6 as targets of myopic axial elongation of mouse eyes. Nat Commun 2022; 13:5859. [PMID: 36216837 PMCID: PMC9550863 DOI: 10.1038/s41467-022-33605-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/23/2022] [Indexed: 11/09/2022] Open
Abstract
Axial length is the primary determinant of eye size, and it is elongated in myopia. However, the underlying mechanism of the onset and progression of axial elongation remain unclear. Here, we show that endoplasmic reticulum (ER) stress in sclera is an essential regulator of axial elongation in myopia development through activation of both PERK and ATF6 axis followed by scleral collagen remodeling. Mice with lens-induced myopia (LIM) showed ER stress in sclera. Pharmacological interventions for ER stress could induce or inhibit myopia progression. LIM activated all IRE1, PERK and ATF6 axis, and pharmacological inhibition of both PERK and ATF6 suppressed myopia progression, which was confirmed by knocking down above two genes via CRISPR/Cas9 system. LIM dramatically changed the expression of scleral collagen genes responsible for ER stress. Furthermore, collagen fiber thinning and expression of dysregulated collagens in LIM were ameliorated by 4-PBA administration. We demonstrate that scleral ER stress and PERK/ATF6 pathway controls axial elongation during the myopia development in vivo model and 4-PBA eye drop is promising drug for myopia suppression/treatment.
Collapse
Affiliation(s)
- Shin-Ichi Ikeda
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan. .,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Xiaoyan Jiang
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yukihiro Miwa
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Naho Serizawa
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Heonuk Jeong
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kiwako Mori
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiromitsu Kunimi
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Nobuhiro Ozawa
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Chiho Shoda
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Mari Ibuki
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hidemasa Torii
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan. .,Tsubota Laboratory, Inc., 34 Shinanomachi, Shinjuku-ku, Tokyo, 160-0016, Japan.
| |
Collapse
|
6
|
Sauriasari R, Safitri DD, Azmi NU. Current updates on protein as biomarkers for diabetic kidney disease: a systematic review. Ther Adv Endocrinol Metab 2021; 12:20420188211049612. [PMID: 34721837 PMCID: PMC8554552 DOI: 10.1177/20420188211049612] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 09/12/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND In the past decade, researchers have been focused on discovering protein biomarkers for diabetic kidney disease. This paper aims to search for, analyze, and synthesize current updates regarding the development of these efforts. METHODS We systematically searched the ScienceDirect, SpringerLink, and PubMed databases for observational studies of protein biomarkers in patients with diabetes mellitus. We included studies published between January 2018 and April 2020, that were based on a population of patients with type-1 or type-2 diabetes mellitus aged ⩾18 years, with an observational design such as cross-sectional, case-control, or cohort studies. The dependent variable of the research results was in the form of protein biomarkers from urine, plasma, or serum. RESULTS Following the screening process, 20 research articles with available full text met the inclusion criteria. These could be categorized as glomerular biomarkers (ANGPTL4, beta-2 microglobulin, Smad1, and glypican-5); inflammatory biomarkers (MCP-1 and adiponectin); and tubular biomarkers (NGAL, VDBP, megalin, sKlotho, and KIM-1). The development of a panel of biomarkers showed more promising results than those for a single biomarker in diagnosing diabetic kidney disease. CONCLUSION All the biomarkers discussed in this review showed promising results for predicting diabetic kidney disease because they correlate with albuminuria, eGFR, or both. However, of the 11 protein biomarkers, none have prognostic value beyond albuminuria and eGFR.
Collapse
Affiliation(s)
| | | | - Nuriza Ulul Azmi
- Faculty of Pharmacy, Universitas Indonesia, Depok, 16424, Indonesia
| |
Collapse
|
7
|
Zhong B, Ma S, Wang DH. Knockout of TRPV1 Exacerbates Ischemia-reperfusion-induced Renal Inflammation and Injury in Obese Mice. In Vivo 2020; 34:2259-2268. [PMID: 32871748 DOI: 10.21873/invivo.12036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND/AIM Transient receptor potential vanilloid type 1 (TRPV1) has anti-inflammatory properties. The present study aimed to investigate the role of TRPV1 in renal inflammatory responses and tissue injury following renal ischemia-reperfusion (I/R) in diet-induced obese mice. MATERIALS AND METHODS TRPV1 knockout and wild type mice were fed a normal or western diet (WD) for 23 weeks and were then subjected to renal I/R injury. RESULTS TRPV1 knockout mice showed enhanced WD-induced renal macrophage infiltration and collagen deposition. Knocking out TRPV1 exacerbated renal I/R-induced increase of malondialdehyde, interleukin-6, monocyte chemoattractant protein-1, and NF-ĸB in obese mice. Similar results were observed in the expression of phosphorylated Smad1 and Smad2/3. Blockade of calcitonin gene-related peptide (CGRP) receptors with CGRP8-37 worsened the I/R-induced renal inflammation and injury. CONCLUSION Our data indicate that preserving TRPV1 expression and function may prevent renal I/R injury in obesity likely through alleviating inflammatory responses.
Collapse
Affiliation(s)
- Beihua Zhong
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI, U.S.A
| | - Shuangtao Ma
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI, U.S.A
| | - Donna H Wang
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI, U.S.A. .,Neuroscience Program, Michigan State University, East Lansing, MI, U.S.A.,Cell & Molecular Biology Program, Michigan State University, East Lansing, MI, U.S.A
| |
Collapse
|
8
|
Veiga G, Alves B, Perez M, Alcantara LV, Raimundo J, Zambrano L, Encina J, Pereira EC, Bacci M, Murad N, Fonseca F. NGAL and SMAD1 gene expression in the early detection of diabetic nephropathy by liquid biopsy. J Clin Pathol 2020; 73:713-721. [PMID: 32184218 DOI: 10.1136/jclinpath-2020-206494] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Diabetic nephropathy (DN) is a disease that progresses with the slow and progressive decline of the glomerular filtration rate (GFR); the installation of this pathology is silent and one of the major causes of death in patients with diabetes. AIMS To identify new molecular biomarkers for early identification of the onset of DN in patients with type II diabetes mellitus (DM2). We studied the expression profile of the genes; suppressor of mothers against decapentaplegic type 1 (SMAD1), neutrophil gelatinase-associated lipocalin (NGAL) and type IV collagen (COLIV1A) in peripheral blood and urine sediment samples. METHODS Ninety volunteers, 51 with DM2 and 39 healthy, were recruited from the Faculdade de Medicina do ABC outpatient clinic. We conducted an interview and collected anthropometric data, as well as blood and urine samples for biochemical evaluation and real-time PCR amplification of the genes of interest. RESULTS Gene expression data: peripheral blood NGAL (DM2 0.09758±0.1914 vs CTL 0.02293±0.04578), SMAD1 (blood: DM2 0.01102±0.04059* vs CTL 0.0001317±0.0003609; urine: DM2 0.7195±2.344* vs CTL 0.09812±0.4755), there was no significant expression of COLIV1A. These genes demonstrated good sensitivity and specificity in the receiving operating characteristic curve evaluation. CONCLUSION Our data suggest the potential use of NGAL and SMAD1 gene expression in peripheral blood and urine samples as early biomarkers of DN.
Collapse
Affiliation(s)
- Glaucia Veiga
- Centro Universitário Saúde ABC/Faculdade de Medicina do ABC, Santo Andre, Brazil
| | - Beatriz Alves
- Centro Universitário Saúde ABC/Faculdade de Medicina do ABC, Santo Andre, Brazil
| | - Matheus Perez
- Centro Universitário Saúde ABC/Faculdade de Medicina do ABC, Santo Andre, Brazil
| | | | - Joyce Raimundo
- Centro Universitário Saúde ABC/Faculdade de Medicina do ABC, Santo Andre, Brazil
| | - Lysien Zambrano
- Universidad Nacional Autónoma de Honduras, Tegucigalpa, Francisco Morazán, Honduras
| | - Jessica Encina
- Centro Universitário Saúde ABC/Faculdade de Medicina do ABC, Santo Andre, Brazil
| | - Edimar Cristiano Pereira
- Pharmaceutical Sciences Department, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| | - Marcelo Bacci
- Centro Universitário Saúde ABC/Faculdade de Medicina do ABC, Santo Andre, Brazil
| | - Neif Murad
- Centro Universitário Saúde ABC/Faculdade de Medicina do ABC, Santo Andre, Brazil
| | - Fernando Fonseca
- Centro Universitário Saúde ABC/Faculdade de Medicina do ABC, Santo Andre, Brazil.,Pharmaceutical Sciences Department, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| |
Collapse
|
9
|
Perera N, Ritchie RH, Tate M. The Role of Bone Morphogenetic Proteins in Diabetic Complications. ACS Pharmacol Transl Sci 2019; 3:11-20. [PMID: 32259084 DOI: 10.1021/acsptsci.9b00064] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 12/22/2022]
Abstract
The prevalence of diabetes has reached epidemic proportions and is placing a significant burden on healthcare systems globally. Diabetes has a detrimental impact on many organs in the human body, including accelerating the development of micro- and macrovascular complications. Current therapeutic options to treat diabetic complications have their limitations. Importantly, many slow but fail to reverse the progression of diabetic complications. Bone morphogenetic proteins (BMPs) are a highly conserved subgroup of the transforming growth factor β (TGFβ) superfamily, signaling via serine/threonine kinase receptors, that have recently been implicated in glucose homeostasis and insulin resistance in the setting of diabetes. Downstream of the receptors, the signal can be transduced via the canonical Smad-dependent pathway or the noncanonical Smad-independent pathways. BMPs are essential in organ development, tissue homeostasis, and, as expected, disease pathogenesis. In fact, deletion of BMPs can be embryonically lethal or result in severe organ abnormalities. This review outlines the BMP signaling pathway and its relevance to diabetic complications, namely, diabetic nephropathy, diabetes-associated cardiovascular diseases, and diabetic retinopathy. Understanding the complexities of BMP signaling and particularly its tissue-, cellular-, and time-dependent actions will help delineate the underlying pathogenesis of the disease and may ultimately be harnessed in the treatment of diabetes-induced complications. This would replicate progress made in numerous other diseases, including cancer and atherosclerosis.
Collapse
Affiliation(s)
- Nimna Perera
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.,Department of Pharmacology and Department of Diabetes, Monash University, Melbourne, Victoria 3800, Australia.,Department of Pharmacology and Department of Diabetes, Monash University, Melbourne, Victoria 3800, Australia
| | - Mitchel Tate
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.,Department of Pharmacology and Department of Diabetes, Monash University, Melbourne, Victoria 3800, Australia
| |
Collapse
|
10
|
Sakurai A, Ono H, Ochi A, Matsuura M, Yoshimoto S, Kishi S, Murakami T, Tominaga T, Nagai K, Abe H, Doi T. Involvement of Elf3 on Smad3 activation-dependent injuries in podocytes and excretion of urinary exosome in diabetic nephropathy. PLoS One 2019; 14:e0216788. [PMID: 31150422 PMCID: PMC6544199 DOI: 10.1371/journal.pone.0216788] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/29/2019] [Indexed: 01/15/2023] Open
Abstract
Diabetic nephropathy (DN) is among the most serious complications of diabetes mellitus, and often leads to end-stage renal disease ultimately requiring dialysis or renal transplantation. The loss of podocytes has been reported to have a role in the onset and progression of DN. Here, we addressed the activation mechanism of Smad3 signaling in podocytes. Expression of RII and activation of Smad3 were induced by AGE exposure (P<0.05). Reduction of the activation of RII-Smad3 signaling ameliorated podocyte injuries in Smad3-knockout diabetic mice. The bone morphogenetic protein 4 (BMP4) significantly regulated activation of RII-Smad3 signalings (P<0.05). Moreover, the epithelium-specific transcription factor, Elf3was induced by AGE stimulation and, subsequently, upregulated RII expression in cultured podocytes. Induction of Elf3 and activation of RII-Smad3 signaling, leading to a decrease in WT1 expression, were observed in podocytes in diabetic human kidneys. Moreover, AGE treatment induced the secretion of Elf3-containing exosomes from cultured podocytes, which was dependent on the activation of the TGF-β-Smad3 signaling pathway. In addition, exosomal Elf3 protein in urine could be measured only in urinary exosomes from patients with DN. The appearance of urinary exosomal Elf3 protein in patients with DN suggested the existence of irreversible injuries in podocytes. The rate of decline in the estimated Glomerular Filtration Rate (eGFR) after measurement of urinary exosomal Elf3 protein levels in patients with DN (R2 = 0.7259) might be useful as an early non-invasive marker for podocyte injuries in DN.
Collapse
Affiliation(s)
- Akiko Sakurai
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hiroyuki Ono
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Arisa Ochi
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Motokazu Matsuura
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Sakiya Yoshimoto
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Seiji Kishi
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Taichi Murakami
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kojiro Nagai
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hideharu Abe
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
- * E-mail:
| | - Toshio Doi
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
11
|
Chen C, Lin J, Li L, Zhu T, Gao L, Wu W, Liu Q, Ou S. The role of the BMP4/Smad1 signaling pathway in mesangial cell proliferation: A possible mechanism of diabetic nephropathy. Life Sci 2019; 220:106-116. [DOI: 10.1016/j.lfs.2019.01.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 01/18/2023]
|
12
|
Tamaki M, Tominaga T, Fujita Y, Koezuka Y, Ichien G, Murakami T, Kishi S, Yamamoto K, Abe H, Nagai K, Doi T. All-trans retinoic acid suppresses bone morphogenetic protein 4 in mouse diabetic nephropathy through a unique retinoic acid response element. Am J Physiol Endocrinol Metab 2019; 316:E418-E431. [PMID: 30601699 DOI: 10.1152/ajpendo.00218.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetic nephropathy (DN) causes mesangial matrix expansion, which results in glomerulosclerosis and renal failure. Collagen IV (COL4) is a major component of the mesangial matrix that is positively regulated by bone morphogenetic protein 4 (BMP4)/suppressor of mothers against decapentaplegic (Smad1) signaling. Because previous studies showed that retinoids treatment had a beneficial effect on kidney disease, we investigated the therapeutic potential of retinoids in DN, focusing especially on the regulatory mechanism of BMP4. Diabetes was induced with streptozotocin in 12-wk-old male Crl:CD1(ICR) mice, and, 1 mo later, we initiated intraperitoneal injection of all-trans retinoic acid (ATRA) three times weekly. Glomerular matrix expansion, which was associated with increased BMP4, phosphorylated Smad1, and COL4 expression, worsened in diabetic mice at 24 wk of age. ATRA administration alleviated DN and downregulated BMP4, phosopho-Smad1, and COL4. In cultured mouse mesangial cells, treatment with ATRA or a retinoic acid receptor-α (RARα) agonist significantly decreased BMP4 and COL4 expression. Genomic analysis suggested two putative retinoic acid response elements (RAREs) for the mouse Bmp4 gene. Chromatin immunoprecipitation analysis and reporter assays indicated a putative RARE of the Bmp4 gene, located 11,488-11,501 bp upstream of exon 1A and bound to RARα and retinoid X receptor (RXR), which suppressed BMP4 expression after ATRA addition. ATRA suppressed BMP4 via binding of a RARα/RXR heterodimer to a unique RARE, alleviating glomerular matrix expansion in diabetic mice. These findings provide a novel regulatory mechanism for treatment of DN.
Collapse
Affiliation(s)
- Masanori Tamaki
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Yui Fujita
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | | | | | - Taichi Murakami
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Seiji Kishi
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | | | - Hideharu Abe
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Kojiro Nagai
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
| | - Toshio Doi
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School , Tokushima , Japan
- Research Institute for Production Development , Kyoto , Japan
| |
Collapse
|
13
|
Merhi Z. Vitamin D attenuates the effect of advanced glycation end products on anti-Mullerian hormone signaling. Mol Cell Endocrinol 2019; 479:87-92. [PMID: 30253183 DOI: 10.1016/j.mce.2018.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022]
Abstract
Vitamin D3 (1,25-dihydroxyvitamin D3, VD3) in vitro attenuates the effect of the pro-inflammatory advanced glycation end products (AGEs) on steroidogenesis in human granulosa cells (GCs) by downregulating the receptor for AGEs (RAGE). It has been shown that VD3 alone downregulates anti-Mullerian hormone (AMH) type 2 receptor (AMHR-2) gene expression and suppresses AMH-induced SMAD 1/5/8 phosphorylation in granulosa cells. However, the effect of AGEs, in the absence or presence of VD3, on AMH action in GCs has not been studied. Using human GCs, this study showed that human glycated albumin (HGA), an in vitro representative for AGEs, upregulated AMHR-2 mRNA but did not alter AMH mRNA expression levels. VD3 inhibited the HGA-induced increase in AMHR-2 mRNA expression levels. In KGN granulosa cell line, recombinant AMH induced SMAD 1/5/8 phosphorylation. HGA augmented the recombinant AMH-induced SMAD 1/5/8 phosphorylation while the addition of VD3 to HGA attenuated the recombinant AMH-induced SMAD 1/5/8 phosphorylation. Thus, AGEs could potentially affect folliculogenesis as reflected by changes in AMH signaling. These findings have significant implications for women with polycystic ovary syndrome who have significantly elevated serum and ovarian AGEs.
Collapse
Affiliation(s)
- Zaher Merhi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Protective Effect of PACAP on Ischemia/Reperfusion-Induced Kidney Injury of Male and Female Rats: Gender Differences. J Mol Neurosci 2018; 68:408-419. [PMID: 30443839 DOI: 10.1007/s12031-018-1207-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 11/08/2018] [Indexed: 02/07/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that exerts general cytoprotective effects, including protection in different kidney disorders. The aim of our study was to investigate the ischemia/reperfusion-induced kidney injury of male and female rats to confirm the protective effects of PACAP in the kidney and to reveal possible gender differences.Male and female Wistar rats underwent unilateral renal artery clamping followed by 24-h, 48-h, or 14-day reperfusion. PACAP was administered intravenously before arterial clamping in half of the rats. Tubular damage, cytokine expression pattern, oxidative stress marker, antioxidative status and signaling pathways were evaluated using histology, immunohistology, cytokine array, PCR, and Western blot. Tubular damage was significantly less severe in the PACAP-treated male and female rats compared to controls. Results of female animals were significantly better in both treated and untreated groups. Cytokine expression, oxidative stress marker and antioxidative status confirmed the histological results. We also revealed that PACAP counteracted the decreased PKA phosphorylation, influenced the expression of BMP2 and BMP4, and increased the expression of the protein Smad1.We conclude that PACAP is protective in ischemia/reperfusion-induced kidney injury in both sexes, but females had markedly less pronounced injury after ischemia/reperfusion, possibly also involving further protective factors, the investigation of which could have future therapeutic value in treating ischemic kidney injuries.
Collapse
|
15
|
Fujita Y, Tominaga T, Abe H, Kangawa Y, Fukushima N, Ueda O, Jishage KI, Kishi S, Murakami T, Saga Y, Kanwar YS, Nagai K, Doi T. An adjustment in BMP4 function represents a treatment for diabetic nephropathy and podocyte injury. Sci Rep 2018; 8:13011. [PMID: 30158674 PMCID: PMC6115362 DOI: 10.1038/s41598-018-31464-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/09/2018] [Indexed: 01/03/2023] Open
Abstract
Podocyte injury has been proposed to play an important role in diabetic nephropathy; however, its pathological mechanism remains unclear. We have shown that bone morphogenetic protein 4 (BMP4) signaling leads to the glomerular changes characteristic of this disorder. To analyze the molecular mechanism of podocyte injury, the effect of BMP4 was investigated using streptozotocin (STZ)-induced, Bmp4 heterozygous knockout (Bmp4+/−) and podocyte-specific Bmp4 knockout mice. Mice with STZ-induced diabetes exhibited glomerular matrix hyperplasia and decreased numbers of podocyte nucleus-specific WT1-positive cells. The number of podocytes and proteinuria were improved in both diabetic Bmp4 knockout mouse models compared to the effects observed in the control mice. The effect of BMP4 overexpression on Bmp4-induced or podocyte-specific transgenic mice was examined. Tamoxifen-induced Bmp4-overexpressing mice exhibited mesangial matrix expansion and decreased numbers of WT1-positive cells. Podocyte-specific Bmp4-overexpressing mice displayed increased kidney BMP4 expression and mesangial matrix expansion but decreased nephrin expression and numbers of WT1-positive cells. Both lines of Bmp4-overexpressing mice exhibited increased albuminuria. In cultured podocytes, BMP4 increased phospho-p38 levels. BMP4 decreased nephrin expression but increased cleaved caspase-3 levels. p38 suppression inhibited caspase-3 activation. Apoptosis was confirmed in STZ-diabetic glomeruli and Bmp4-overexpressing mice. Bmp4 +/− mice with diabetes displayed reduced apoptosis. Based on these data, the BMP4 signaling pathway plays important roles in the development of both podocyte injury and mesangial matrix expansion in diabetic nephropathy.
Collapse
Affiliation(s)
- Yui Fujita
- Department of Nephrology, Graduate School of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Graduate School of Biomedical Science, Tokushima University, Tokushima, Japan.
| | - Hideharu Abe
- Department of Nephrology, Graduate School of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Yumi Kangawa
- Department of Nephrology, Graduate School of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Naoshi Fukushima
- Research Division, Fuji Gotemba Research Labs, Chugai Pharmaceutical Co., Ltd, Shizuoka, Japan
| | - Otoya Ueda
- Research Division, Fuji Gotemba Research Labs, Chugai Pharmaceutical Co., Ltd, Shizuoka, Japan
| | - Kou-Ichi Jishage
- Research Division, Fuji Gotemba Research Labs, Chugai Pharmaceutical Co., Ltd, Shizuoka, Japan.,Chugai Research Institute for Medical Science Inc., Shizuoka, Japan
| | - Seiji Kishi
- Department of Nephrology, Graduate School of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Taichi Murakami
- Department of Nephrology, Graduate School of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Yumiko Saga
- Division of Mammalian Development, Genetic Strains Research Center, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Yashpal S Kanwar
- Department of Pathology & Medicine-Nephrology, FSM, Northwestern University, Chicago, Illinois, 60611, USA
| | - Kojiro Nagai
- Department of Nephrology, Graduate School of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Toshio Doi
- Department of Nephrology, Graduate School of Biomedical Science, Tokushima University, Tokushima, Japan
| |
Collapse
|
16
|
Novel Interplay Between Smad1 and Smad3 Phosphorylation via AGE Regulates the Progression of Diabetic Nephropathy. Sci Rep 2018; 8:10548. [PMID: 30002389 PMCID: PMC6043613 DOI: 10.1038/s41598-018-28439-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/18/2018] [Indexed: 11/08/2022] Open
Abstract
Diabetic nephropathy (DN) is the major cause of end-stage renal failure and is associated with increased morbidity and mortality compared with other causes of renal diseases. We previously found that Smad1 plays a critical role in the development of DN both in vitro and in vivo. However, functional interaction between Smad1 and Smad3 signaling in DN is unclear. Here, we addressed the molecular interplay between Smad1 and Smad3 signaling under a diabetic condition by using Smad3-knockout diabetic mice. Extracellular matrix (ECM) protein overexpression and Smad1 activation were observed in the glomeruli of db/db mice but were suppressed in the glomeruli of Smad3+/-; db/db mice. Smad3 activation enhanced the phosphorylation of Smad1 C-terminal domain but decreased the phosphorylation of linker domain, thus regulating Smad1 activation in advanced glycation end product-treated mesangial cells (MCs). However, forced phosphorylation of the Smad1 linker domain did not affect Smad3 activation in MCs. Phosphorylation of the Smad1 linker domain increased in Smad3+/-; db/db mice and probucol-treated db/db mice, which was consistent with the attenuation of ECM overproduction. These results indicate that Smad3 expression and activation or probucol treatment alters Smad1 phosphorylation, thus suggesting new molecular mechanisms underlying DN development and progression.
Collapse
|
17
|
Doi T, Moriya T, Fujita Y, Minagawa N, Usami M, Sasaki T, Abe H, Kishi S, Murakami T, Ouchi M, Ichien G, Yamamoto K, Ikeda H, Koezuka Y, Takamatsu N, Shima K, Mauer M, Nagai K, Tominaga T. Urinary IgG4 and Smad1 Are Specific Biomarkers for Renal Structural and Functional Changes in Early Stages of Diabetic Nephropathy. Diabetes 2018; 67:986-993. [PMID: 29490904 DOI: 10.2337/db17-1043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/16/2018] [Indexed: 11/13/2022]
Abstract
Diabetic nephropathy (DN) is the major cause of end-stage kidney disease, but early biomarkers of DN risk are limited. Herein we examine urinary IgG4 and Smad1 as additional early DN biomarkers. We recruited 815 patients with type 2 diabetes; 554 patients fulfilled the criteria of an estimated glomerular filtration rate (eGFR) >60 mL/min and no macroalbuminuria at baseline, with follow-up for 5 years. Patients without macroalbuminuria were also recruited for renal biopsies. Urinary IgG4 and Smad1 were determined by enzyme-linked immunoassays using specific antibodies. The specificity, sensitivity, and reproducibility were confirmed for each assay. Increased urinary IgG4 was significantly associated with lower eGFR. The level of urinary IgG4 also significantly correlated with surface density of peripheral glomerular basement membrane (Sv PGBM/Glom), whereas Smad1 was associated with the degree of mesangial expansion-both classic pathological findings in DN. Baseline eGFR did not differ between any groups; however, increases in both urinary IgG4 and Smad1 levels at baseline significantly predicted later development of eGFR decline in patients without macroalbuminuria. These data suggest that urinary IgG4 and Smad1 at relatively early stages of DN reflect underlying DN lesions and are relevant to later clinical outcomes.
Collapse
Affiliation(s)
- Toshio Doi
- Department of Nephrology, Graduate School of Medical Science, Tokushima University, Tokushima, Japan
| | - Tatsumi Moriya
- Health Care Center, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Yui Fujita
- Department of Nephrology, Graduate School of Medical Science, Tokushima University, Tokushima, Japan
| | | | | | | | - Hideharu Abe
- Department of Nephrology, Graduate School of Medical Science, Tokushima University, Tokushima, Japan
| | - Seiji Kishi
- Department of Nephrology, Graduate School of Medical Science, Tokushima University, Tokushima, Japan
| | - Taichi Murakami
- Department of Nephrology, Graduate School of Medical Science, Tokushima University, Tokushima, Japan
| | - Motoshi Ouchi
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | | | | | | | | | | | | | - Michael Mauer
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Kojiro Nagai
- Department of Nephrology, Graduate School of Medical Science, Tokushima University, Tokushima, Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Graduate School of Medical Science, Tokushima University, Tokushima, Japan
| |
Collapse
|
18
|
Thomsen LH, Fog-Tonnesen M, Nielsen Fink L, Norlin J, García de Vinuesa A, Hansen TK, de Heer E, Ten Dijke P, Rosendahl A. Disparate phospho-Smad2 levels in advanced type 2 diabetes patients with diabetic nephropathy and early experimental db/db mouse model. Ren Fail 2018; 39:629-642. [PMID: 28805484 PMCID: PMC6446227 DOI: 10.1080/0886022x.2017.1361837] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Uncontrolled activation of transforming growth factor beta (TGF-β) family members is hypothesized to participate in type 2 diabetes (T2D) dependent diabetic nephropathy (DN). We evaluated and compared downstream activation of the Smad2-signaling pathway in kidney samples from T2D patients to kidneys from the T2D model of leptin receptor deficient db/db mouse. Furthermore, expression of TGF-β family members was evaluated to elucidate molecular mechanisms in the mouse model. Kidney samples from patients with advanced stages of DN showed elevated pSmad2 staining whereas db/db mouse kidneys surprisingly showed a decrease in pSmad2 in the tubular compartment. Structurally, kidney tissue showed dilated tubules and expanded glomeruli, but no clear fibrotic pattern was found in the diabetic mice. Selective TGF-β family members were up-regulated at the mRNA level. Antagonists of bone morphogenetic protein (BMP) ligands, such as Gremlin1, USAG1 and Sclerostin, were strongly up-regulated suggesting a dampening effect on BMP pathways. Together, these results indicate a lack of translation from T2D patient kidneys to the db/db model with regards to Smad signaling pathway. It is plausible that a strong up-regulation of BMP antagonizing factors account for the lack of Smad1/5/8 activation, in spite of increased expression of several BMP members.
Collapse
Affiliation(s)
- Lise Høj Thomsen
- a Department of Diabetes Complications Research , Novo Nordisk A/S , Måløv , Denmark.,b Department of Endocrinology and Internal Medicine , Aarhus University Hospital , Aarhus , Denmark
| | - Morten Fog-Tonnesen
- a Department of Diabetes Complications Research , Novo Nordisk A/S , Måløv , Denmark
| | - Lisbeth Nielsen Fink
- a Department of Diabetes Complications Research , Novo Nordisk A/S , Måløv , Denmark
| | - Jenny Norlin
- c Department of Incretin & Obesity Pharmacology , Novo Nordisk A/S , Måløv , Denmark
| | - Amaya García de Vinuesa
- d Department of Molecular Cell Biology , Cancer Genomics Centre Netherlands, Leiden University Medical Center , Leiden , The Netherlands
| | - Troels Krarup Hansen
- b Department of Endocrinology and Internal Medicine , Aarhus University Hospital , Aarhus , Denmark
| | - Emile de Heer
- e Department of Pathology , Leiden University Medical Center , Leiden , The Netherlands
| | - Peter Ten Dijke
- d Department of Molecular Cell Biology , Cancer Genomics Centre Netherlands, Leiden University Medical Center , Leiden , The Netherlands
| | - Alexander Rosendahl
- a Department of Diabetes Complications Research , Novo Nordisk A/S , Måløv , Denmark
| |
Collapse
|
19
|
Halperin Kuhns VL, Pluznick JL. Novel differences in renal gene expression in a diet-induced obesity model. Am J Physiol Renal Physiol 2017; 314:F517-F530. [PMID: 29141937 DOI: 10.1152/ajprenal.00345.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Obesity is a significant risk factor for both chronic kidney disease and end-stage renal disease. To better understand disease development, we sought to identify novel genes differentially expressed early in disease progression. We first confirmed that mice fed a high-fat (HF) diet exhibit early signs of renal injury including hyperfiltration. We then performed RNA-Seq using renal cortex RNA from C57BL6/J male mice fed either HF or control (Ctrl) diet. We identified 1,134 genes differentially expressed in the cortex on HF vs. Ctrl, of which 31 genes were selected for follow-up analysis. This included the 9 most upregulated, the 11 most downregulated, and 11 genes of interest (primarily sensory receptors and G proteins). Quantitative (q)RT-PCR for these 31 genes was performed on additional male renal cortex and medulla samples, and 11 genes (including all 9 upregulated genes) were selected for further study based on qRT-PCR. We then examined expression of these 11 genes in Ctrl and HF male heart and liver samples, which demonstrated that these changes are relatively specific to the renal cortex. These 11 genes were also examined in female renal cortex, where we found that the expression changes seen in males on a HF diet are not replicated in females, even when the females are started on the diet sooner to match weight gain of the males. In sum, these data demonstrate that in a HF-diet model of early disease, novel transcriptional changes occur that are both sex specific and specific to the renal cortex.
Collapse
Affiliation(s)
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
20
|
Chaudhari S, Li W, Wang Y, Jiang H, Ma Y, Davis ME, Zuckerman JE, Ma R. Store-operated calcium entry suppressed the TGF-β1/Smad3 signaling pathway in glomerular mesangial cells. Am J Physiol Renal Physiol 2017. [PMID: 28637791 DOI: 10.1152/ajprenal.00483.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Our previous study demonstrated that the abundance of extracellular matrix proteins was suppressed by store-operated Ca2+ entry (SOCE) in mesangial cells (MCs). The present study was conducted to investigate the underlying mechanism focused on the transforming growth factor-β1 (TGF-β1)/Smad3 pathway, a critical pathway for ECM expansion in diabetic kidneys. We hypothesized that SOCE suppressed ECM protein expression by inhibiting this pathway in MCs. In cultured human MCs, we observed that TGF-β1 (5 ng/ml for 15 h) significantly increased Smad3 phosphorylation, as evaluated by immunoblot. However, this response was markedly inhibited by thapsigargin (1 µM), a classical activator of store-operated Ca2+ channels. Consistently, both immunocytochemistry and immunoblot showed that TGF-β1 significantly increased nuclear translocation of Smad3, which was prevented by pretreatment with thapsigargin. Importantly, the thapsigargin effect was reversed by lanthanum (La3+; 5 µM) and GSK-7975A (10 µM), both of which are selective blockers of store-operated Ca2+ channels. Furthermore, knockdown of Orai1, the pore-forming subunit of the store-operated Ca2+ channels, significantly augmented TGF-β1-induced Smad3 phosphorylation. Overexpression of Orai1 augmented the inhibitory effect of thapsigargin on TGF-β1-induced phosphorylation of Smad3. In agreement with the data from cultured MCs, in vivo knockdown of Orai1 specific to MCs using a targeted nanoparticle small interfering RNA delivery system resulted in a marked increase in abundance of phosphorylated Smad3 and in nuclear translocation of Smad3 in the glomerulus of mice. Taken together, our results indicate that SOCE in MCs negatively regulates the TGF-β1/Smad3 signaling pathway.
Collapse
Affiliation(s)
- Sarika Chaudhari
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas
| | - Weizu Li
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas.,Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Yanxia Wang
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas
| | - Hui Jiang
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas.,First Hospital Affiliated to Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Yuhong Ma
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas.,Department of Clinical Medicine, Wanna Medical College, Wuhu, Anhui, China; and
| | - Mark E Davis
- Department of Chemical Engineering, California Institute of Technology, Pasadena, California
| | - Jonathan E Zuckerman
- Department of Chemical Engineering, California Institute of Technology, Pasadena, California
| | - Rong Ma
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, Fort Worth, Texas;
| |
Collapse
|
21
|
Wu P, Ren Y, Ma Y, Wang Y, Jiang H, Chaudhari S, Davis ME, Zuckerman JE, Ma R. Negative regulation of Smad1 pathway and collagen IV expression by store-operated Ca 2+ entry in glomerular mesangial cells. Am J Physiol Renal Physiol 2017; 312:F1090-F1100. [PMID: 28298362 DOI: 10.1152/ajprenal.00642.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/08/2017] [Accepted: 03/09/2017] [Indexed: 12/13/2022] Open
Abstract
Collagen IV (Col IV) is a major component of expanded glomerular extracellular matrix in diabetic nephropathy and Smad1 is a key molecule regulating Col IV expression in mesangial cells (MCs). The present study was conducted to determine if Smad1 pathway and Col IV protein abundance were regulated by store-operated Ca2+ entry (SOCE). In cultured human MCs, pharmacological inhibition of SOCE significantly increased the total amount of Smad1 protein. Activation of SOCE blunted high-glucose-increased Smad1 protein content. Treatment of human MCs with ANG II at 1 µM for 15 min, high glucose for 3 days, or TGF-β1 at 5 ng/ml for 30 min increased the level of phosphorylated Smad1. However, the phosphorylation of Smad1 by those stimuli was significantly attenuated by activation of SOCE. Knocking down Smad1 reduced, but expressing Smad1 increased, the amount of Col IV protein. Furthermore, activation of SOCE significantly attenuated high-glucose-induced Col IV protein production, and blockade of SOCE substantially increased the abundance of Col IV. To further verify those in vitro findings, we downregulated SOCE specifically in MCs in mice using small-interfering RNA (siRNA) against Orai1 (the channel protein mediating SOCE) delivered by the targeted nanoparticle delivery system. Immunohistochemical examinations showed that expression of both Smad1 and Col IV proteins was significantly greater in the glomeruli with positively transfected Orai1 siRNA compared with the glomeruli from the mice without Orai1 siRNA treatment. Taken together, our results indicate that SOCE negatively regulates the Smad1 signaling pathway and inhibits Col IV protein production in MCs.
Collapse
Affiliation(s)
- Peiwen Wu
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas.,Department of Endocrinology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, Peoples Republic of China
| | - Yuezhong Ren
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas.,Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, Zhejiang, China
| | - Yuhong Ma
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas.,Department of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Yanxia Wang
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas
| | - Hui Jiang
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas.,The First Affiliated Hospital to Anhui University of Traditional Chinese Medicine, Hefei, China; and
| | - Sarika Chaudhari
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas
| | - Mark E Davis
- Chemical Engineering, California Institute of Technology, Pasadena, California
| | | | - Rong Ma
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas;
| |
Collapse
|
22
|
Mi X, Tang W, Chen X, Liu F, Tang X. Mitofusin 2 attenuates the histone acetylation at collagen IV promoter in diabetic nephropathy. J Mol Endocrinol 2016; 57:233-249. [PMID: 27997345 DOI: 10.1530/jme-16-0031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/03/2016] [Indexed: 12/15/2022]
Abstract
Extracellular matrix (ECM) increase in diabetic nephropathy (DN) is closely related to mitochondrial dysfunction. The mechanism of protective function of mitofusin 2 (Mfn2) for mitochondria remains largely unknown. In this study, the molecular mechanisms for the effect of Mfn2 on mitochondria and subsequent collagen IV expression in DN were investigated. Ras-binding-deficient mitofusin 2 (Mfn2-Ras(Δ)) were overexpressed in rat glomerular mesangial cells, and then the cells were detected for mitochondrial morphology, cellular reactive oxygen species (ROS), mRNA and protein expression of collagen IV with advanced glycation end-product (AGE) stimulation. Preliminary results reveal that the mitochondrial dysfunction and the increased synthesis of collagen IV after AGE stimulation were reverted by Mfn2-Ras(Δ) overexpression. Bioinformatical computations were performed to search transcriptional factor motifs in the promoter region of collagen IV. Three specific regions for TFAP2A binding were identified, followed by validation with chromatin immunoprecipitation experiments. Knocking down TFAP2A significantly decreased the TF binding in the first two regions and the gene expression of collagen IV. Furthermore, results reveal that Mfn2-Ras(Δ) overexpression significantly mitigated TFAP2A binding and also reverted the histone acetylation at Regions 1 and 2 after AGE stimulation. In streptozotocin-induced diabetic rats, Mfn2-Ras(Δ) overexpression also ameliorated glomerular mesangial lesions with decreased collagen IV expression, accompanied by decreased acetylation and TFAP2A binding at Region 1. In conclusion, this study highlights the pathway by which mitochondria affect the histone acetylation of gene promoter and provides a new potential therapy approach for DN.
Collapse
Affiliation(s)
- Xuhua Mi
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Wanxin Tang
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Xiaolei Chen
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Fei Liu
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Xiaohong Tang
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Inada A, Inada O, Fujii NL, Nagafuchi S, Katsuta H, Yasunami Y, Matsubara T, Arai H, Fukatsu A, Nabeshima YI. Adjusting the 17β-Estradiol-to-Androgen Ratio Ameliorates Diabetic Nephropathy. J Am Soc Nephrol 2016; 27:3035-3050. [PMID: 26940099 PMCID: PMC5042662 DOI: 10.1681/asn.2015070741] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/14/2016] [Indexed: 12/28/2022] Open
Abstract
Diabetes is manifested predominantly in males in experimental models, and compelling evidence suggests that 17β-estradiol (E2) supplementation improves hyperglycemia in humans. We previously generated a severely diabetic transgenic (Tg) mouse model by β-cell–specific overexpression of inducible cAMP early repressor (ICER) and found that male but not female ICER-Tg mice exhibit sustained hyperglycemia and develop major clinical and pathologic features of human diabetic nephropathy (DN). Thus, we hypothesized that differences in circulating hormone levels have a key role in determining susceptibility to diabetes. Here, we examined whether DN in male ICER-Tg mice is rescued by adjusting the androgen-to-E2 ratio to approximate that in normoglycemic female ICER-Tg mice. We treated hyperglycemic male ICER-Tg mice with orchiectomy (ORX), E2 pellet implantation, or both. E2 pellet implantation at an early stage of DN with or without ORX caused a rapid drop in blood glucose and a dramatic increase in β-cell number, and it markedly inhibited DN progression [namely, E2 reduced glomerulosclerosis, collagen 4 deposition and albuminuria, and prevented hyperfiltration]. Furthermore, E2 pellet implantation was more effective than ORX alone and induced a remarkable improvement, even when initiated at advanced-stage DN. In contrast, induction of normoglycemia by islet transplant in ICER-Tg mice eliminated albuminuria but was less effective than E2 + ORX in reducing glomerulosclerosis, collagen 4 deposition, and hyperfiltration. These findings indicate that E2 treatment is effective, even after establishment of DN, whereas glucose normalization alone does not improve sclerotic lesions. We propose that E2 intervention is a potential therapeutic option for DN.
Collapse
Affiliation(s)
- Akari Inada
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Kobe, Japan;
| | - Oogi Inada
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Kobe, Japan; Diabetes and Genes, Advanced Medical Initiatives and
| | - Nobuharu L Fujii
- Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Seiho Nagafuchi
- Department of Medical Science and Technology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hitoshi Katsuta
- Department of Medical Science and Technology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Takeshi Matsubara
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidenori Arai
- Center for Gerontology and Social Science, National Center for Geriatrics and Gerontology, Obu, Japan; and
| | | | - Yo-Ichi Nabeshima
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Kobe, Japan
| |
Collapse
|
24
|
Conditional Deletion of Smad1 Ameliorates Glomerular Injury in Progressive Glomerulonephritis. Sci Rep 2016; 6:31216. [PMID: 27492138 PMCID: PMC4974558 DOI: 10.1038/srep31216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/14/2016] [Indexed: 12/02/2022] Open
Abstract
Matrix expansion and cell proliferation are concomitantly observed in various glomerular injuries. However, the molecular mechanisms responsible for these changes have not been fully elucidated. We have reported that Smad1 is a key signalling molecule that regulates the transcription of type IV collagen (Col4) in mesangial matrix expansion and is thereby involved in glomerular injury in an acute model of glomerulonephritis. In this study, we addressed the role of Smad1 signalling in accelerated nephrotoxic nephritis (NTN), a model of progressive glomerulonephritis, using conditional deletion of Smad1 in Rosa26CreERT2 mice (Smad1-CKO). Mesangial matrix expansion in the Smad1-CKO mice with NTN was significantly inhibited compared with that in wild type mice with NTN, which was consistent with the decrease in Col4 expression level. On the other hand, STAT3 activation and cell proliferation were not influenced by Smad1 deletion in the NTN model. Therefore, we investigated another factor that activates cell proliferation in the absence of Smad1. Id2 induced VEGF secretion and subsequent STAT3 activation, independently of Smad1 expression in mouse mesangial cells. Here we show that Smad1 plays an important role in the development of glomerular injury without affecting cell proliferation, in progressive glomerulonephritis.
Collapse
|
25
|
Villacorte M, Delmarcelle AS, Lernoux M, Bouquet M, Lemoine P, Bolsée J, Umans L, de Sousa Lopes SC, Van Der Smissen P, Sasaki T, Bommer G, Henriet P, Refetoff S, Lemaigre FP, Zwijsen A, Courtoy PJ, Pierreux CE. Thyroid follicle development requires Smad1/5- and endothelial cell-dependent basement membrane assembly. Development 2016; 143:1958-70. [PMID: 27068110 DOI: 10.1242/dev.134171] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/01/2016] [Indexed: 12/12/2022]
Abstract
Thyroid follicles, the functional units of the thyroid gland, are delineated by a monolayer of thyrocytes resting on a continuous basement membrane. The developmental mechanisms of folliculogenesis, whereby follicles are formed by the reorganization of a non-structured mass of non-polarized epithelial cells, are largely unknown. Here we show that assembly of the epithelial basement membrane is crucial for folliculogenesis and is controlled by endothelial cell invasion and by BMP-Smad signaling in thyrocytes. Thyroid-specific Smad1 and Smad5 double-knockout (Smad1/5(dKO)) mice displayed growth retardation, hypothyroidism and defective follicular architecture. In Smad1/5(dKO) embryonic thyroids, epithelial cells remained associated in large clusters and formed small follicles. Although similar follicular defects are found in Vegfa knockout (Vegfa(KO)) thyroids, Smad1/5(dKO) thyroids had normal endothelial cell density yet impaired endothelial differentiation. Interestingly, both Vegfa(KO) and Smad1/5(dKO) thyroids displayed impaired basement membrane assembly. Furthermore, conditioned medium (CM) from embryonic endothelial progenitor cells (eEPCs) rescued the folliculogenesis defects of both Smad1/5(dKO) and Vegfa(KO) thyroids. Laminin α1, β1 and γ1, abundantly released by eEPCs into CM, were crucial for folliculogenesis. Thus, epithelial Smad signaling and endothelial cell invasion promote folliculogenesis via assembly of the basement membrane.
Collapse
Affiliation(s)
- Mylah Villacorte
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | | | - Manon Lernoux
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Mahé Bouquet
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Pascale Lemoine
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Jennifer Bolsée
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Lieve Umans
- VIB Center for the Biology of Disease, KU Leuven, 3000 Leuven, Belgium Laboratory of Molecular Biology (Celgen), Stem Cell Biology and Embryology, KU Leuven, 2333 Leuven, Belgium
| | | | | | - Takako Sasaki
- Department of Matrix Medicine, Faculty of Medicine, Oita University, 879-5593 Oita, Japan
| | - Guido Bommer
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Patrick Henriet
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Samuel Refetoff
- Department of Medicine, Pediatrics and Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Frédéric P Lemaigre
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | - An Zwijsen
- VIB Center for the Biology of Disease, KU Leuven, 3000 Leuven, Belgium Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Pierre J Courtoy
- de Duve Institute and Université Catholique de Louvain, 1200 Brussels, Belgium
| | | |
Collapse
|
26
|
Matsubara T, Araki M, Abe H, Ueda O, Jishage KI, Mima A, Goto C, Tominaga T, Kinosaki M, Kishi S, Nagai K, Iehara N, Fukushima N, Kita T, Arai H, Doi T. Bone Morphogenetic Protein 4 and Smad1 Mediate Extracellular Matrix Production in the Development of Diabetic Nephropathy. Diabetes 2015; 64:2978-90. [PMID: 25995358 DOI: 10.2337/db14-0893] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 04/12/2015] [Indexed: 01/20/2023]
Abstract
Diabetic nephropathy is the leading cause of end-stage renal disease. It is pathologically characterized by the accumulation of extracellular matrix in the mesangium, of which the main component is α1/α2 type IV collagen (Col4a1/a2). Recently, we identified Smad1 as a direct regulator of Col4a1/a2 under diabetic conditions in vitro. Here, we demonstrate that Smad1 plays a key role in diabetic nephropathy through bone morphogenetic protein 4 (BMP4) in vivo. Smad1-overexpressing mice (Smad1-Tg) were established, and diabetes was induced by streptozotocin. Nondiabetic Smad1-Tg did not exhibit histological changes in the kidney; however, the induction of diabetes resulted in an ∼1.5-fold greater mesangial expansion, consistent with an increase in glomerular phosphorylated Smad1. To address regulatory factors of Smad1, we determined that BMP4 and its receptor are increased in diabetic glomeruli and that diabetic Smad1-Tg and wild-type mice treated with a BMP4-neutralizing antibody exhibit decreased Smad1 phosphorylation and ∼40% less mesangial expansion than those treated with control IgG. Furthermore, heterozygous Smad1 knockout mice exhibit attenuated mesangial expansion in the diabetic condition. The data indicate that BMP4/Smad1 signaling is a critical cascade for the progression of mesangial expansion and that blocking this signal could be a novel therapeutic strategy for diabetic nephropathy.
Collapse
Affiliation(s)
| | - Makoto Araki
- Department of Nephrology, Kyoto University, Kyoto, Japan
| | - Hideharu Abe
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | - Otoya Ueda
- Chugai Pharmaceutical Co., Ltd., Shizuoka, Japan
| | | | - Akira Mima
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | - Chisato Goto
- Chugai Research Institute for Medical Science, Inc., Shizuoka, Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | | | - Seiji Kishi
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | - Kojiro Nagai
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | | | | | - Toru Kita
- Kobe City Medical Center General Hospital, Kyoto, Japan
| | - Hidenori Arai
- National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Toshio Doi
- Department of Nephrology, Tokushima University, Tokushima, Japan
| |
Collapse
|
27
|
Korish AA, Abdel Gader AG, Korashy HM, Al-Drees AM, Alhaider AA, Arafah MM. Camel milk attenuates the biochemical and morphological features of diabetic nephropathy: inhibition of Smad1 and collagen type IV synthesis. Chem Biol Interact 2015; 229:100-8. [PMID: 25617480 DOI: 10.1016/j.cbi.2015.01.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 11/27/2014] [Accepted: 01/07/2015] [Indexed: 12/11/2022]
Abstract
Diabetic nephropathy (DN) is a common microvascular complication of diabetes mellitus (DM) that worsens its morbidity and mortality. There is evidence that camel milk (CM) improves the glycemic control in DM but its effect on the renal complications especially the DN remains unclear. Thus the current study aimed to characterize the effects of CM treatment on streptozotocin (STZ)-induced DN. Using STZ-induced diabetes, we investigated the effect of CM treatment on kidney function, proteinuria, renal Smad1, collagen type IV (Col4), blood glucose, insulin resistance (IR), lipid peroxidation, the antioxidant superoxide dismutase (SOD), catalase (CAT) and glutathione (GSH). In addition renal morphology was also examined. The current results showed that rats with untreated diabetes exhibited marked hyperglycemia, IR, high serum urea and creatinine levels, excessive proteinuria, increased renal Smad1 and Col4, glomerular expansion, and extracellular matrix deposition. There was also increased lipid peroxidation products, decreased antioxidant enzyme activity and GSH levels. Camel milk treatment decreased blood glucose, IR, and lipid peroxidation. Superoxide dismutase and CAT expression, CAT activity, and GSH levels were increased. The renoprotective effects of CM were demonstrated by the decreased serum urea and creatinine, proteinuria, Smad1, Col4, and preserved normal tubulo-glomerular morphology. In conclusion, beside its hypoglycemic action, CM attenuates the early changes of DN, decreased renal Smad1 and Col4. This could be attributed to a primary action on the glomerular mesangial cells, or secondarily to the hypoglycemic and antioxidant effects of CM. The protective effects of CM against DN support its use as an adjuvant anti-diabetes therapy.
Collapse
Affiliation(s)
- Aida A Korish
- Physiology Department (29), College of Medicine, King Saud University, PO Box 2925, Riyadh 11461, Riyadh, Saudi Arabia
| | - Abdel Galil Abdel Gader
- Physiology Department (29), College of Medicine, King Saud University, PO Box 2925, Riyadh 11461, Riyadh, Saudi Arabia
| | - Hesham M Korashy
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia.
| | - Abdul Majeed Al-Drees
- Physiology Department (29), College of Medicine, King Saud University, PO Box 2925, Riyadh 11461, Riyadh, Saudi Arabia
| | - Abdulqader A Alhaider
- Physiology Department (29), College of Medicine, King Saud University, PO Box 2925, Riyadh 11461, Riyadh, Saudi Arabia; Camel Biomedical Research Unit, College of Pharmacy and Medicine, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia
| | - Maha M Arafah
- Pathology Department, College of Medicine, King Saud University, PO Box 2925, Riyadh 11461, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Jiao L, Stolzenberg-Solomon R, Zimmerman TP, Duan Z, Chen L, Kahle L, Risch A, Subar AF, Cross AJ, Hollenbeck A, Vlassara H, Striker G, Sinha R. Dietary consumption of advanced glycation end products and pancreatic cancer in the prospective NIH-AARP Diet and Health Study. Am J Clin Nutr 2015; 101:126-34. [PMID: 25527756 PMCID: PMC4266882 DOI: 10.3945/ajcn.114.098061] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Advanced glycation end products (AGEs) are a heterogeneous group of compounds present in uncooked foods as well as in foods cooked at high temperatures. AGEs have been associated with insulin resistance, oxidative stress, and chronic inflammation in patients with diabetes. Dietary AGEs are an important contributor to the AGE pool in the body. N(ϵ)-(carboxymethyl)lysine (CML) AGE is one of the major biologically and chemically well-characterized AGE markers. The consumption of red meat, which is CML-AGE rich, has been positively associated with pancreatic cancer in men. OBJECTIVES With the use of a published food CML-AGE database, we estimated the consumption of CML AGE in the prospective NIH-AARP Diet and Health Study and evaluated the association between CML-AGE consumption and pancreatic cancer and the mediating effect of CML AGE on the association between red meat consumption and pancreatic cancer. DESIGN Multivariate Cox proportional hazard regression models were used to estimate HRs and 95% CIs for pancreatic cancer. RESULTS During an average of 10.5 y of follow-up, we identified 2193 pancreatic cancer cases (1407 men and 786 women) from 528,251 subjects. With the comparison of subjects in the fifth and the first quintiles of CML-AGE consumption, we observed increased pancreatic cancer risk in men (HR: 1.43; 95% CI: 1.06, 1.93, P-trend = 0.003) but not women (HR: 1.14; 95% CI: 0.76, 1.72, P-trend = 0.42). Men in the highest quintile of red meat consumption had higher risk of pancreatic cancer (HR: 1.35; 95% CI: 1.07, 1.70), which attenuated after adjustment for CML-AGE consumption (HR: 1.20; 95% CI: 0.95, 1.53). CONCLUSION Dietary CML-AGE consumption was associated with modestly increased risk of pancreatic cancer in men and may partially explain the positive association between red meat and pancreatic cancer.
Collapse
Affiliation(s)
- Li Jiao
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Rachael Stolzenberg-Solomon
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Thea Palmer Zimmerman
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Zhigang Duan
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Liang Chen
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Lisa Kahle
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Adam Risch
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Amy F Subar
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Amanda J Cross
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Albert Hollenbeck
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Helen Vlassara
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Gary Striker
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| | - Rashmi Sinha
- From the Sections of Gastroenterology and Hepatology (LJ and LC) and Health Services Research (LJ, ZD, and LC), Department of Medicine, Baylor College of Medicine, Houston, TX; the Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics (RS-S and RS) and the Applied Research Program, Division of Cancer Control and Population Sciences (AFS), National Cancer Institute, NIH, Rockville, MD; Westat, Rockville, MD (TPZ); Information Management Services, Rockville, MD (LK and AR); the Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, United Kingdom (AJC); the Division of Experimental Diabetes and Aging, Mount Sinai School of Medicine, New York, NY (HV and GS); and AARP, Washington, DC (AH)
| |
Collapse
|
29
|
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
|
30
|
Li Q, Feng L, Li J, Chen Q. Urinary Smad1 is a new biomarker for diagnosis and evaluating the severity of diabetic nephropathy. Endocrine 2014; 46:83-9. [PMID: 23943254 DOI: 10.1007/s12020-013-0033-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 08/02/2013] [Indexed: 10/26/2022]
Abstract
The aim of this study was to analyze urinary Smad1 level in patients with type 2 diabetes, explore the possibility of Smad1 being a biomarker for early diagnosis and evaluation of severity of diabetic nephropathy, and explore the impact factors affecting urinary Smad1 concentration. In this study, 132 subjects with type 2 diabetes and 50 healthy volunteers were enrolled. Subjects were grouped according to urine albumin to creatinine ratio (ACR) into: normal albumin in urine (NAU), low albumin in urine (LAU), high albumin in urine (HAU), and very high albumin in urine (VHAU). Among those, LAU, HAU, and VHAU were regarded as the diabetic nephropathy group (DN group), NAU was regarded as nondiabetic nephropathy (non-DN group), and the healthy volunteers were the controls. Enzyme-linked immunosorbent assay was used to detect the urinary Smad1 concentration, urinary Smad1 to creatinine ratio (SCR) was used as the standard reference. Compared with non-DN group, SCR of DN group was higher (P < 0.05), while there was no difference between the non-DN group and controls (P > 0.05). There was no significant difference for SCR between LAU and NAU groups (P > 0.05). The SCR was higher in VHAU group than those in HAU and LAU groups, and higher in HAU than that in LAU group (P < 0.05). Pearson correlation analysis showed that SCR measures were positively correlated to ACR, duration and diabetic retinopathy of the disease (r = 0.285, 0.230, 0.202; P = 0.001, 0.008, 0.019, respectively). Multiple linear regression analysis showed that ACR and duration were independent impact factors for SCR (P < 0.05). This is the first known study examining the correlation of Smad1 and DN in clinical practice. It suggested that the urinary Smad1 may be a potential diagnostic parameter for DN and may be used to evaluate the severity of DN. However, it cannot predict those in patients with the earliest DN and low urine albumin concentration. Furthermore, ACR and duration may be independent impact factors for urinary Smad1.
Collapse
Affiliation(s)
- Qiao Li
- Department of Endocrinology, The First Affiliated Hospital, Jinan University, Huangpu Avenue West 613#, Guangzhou, 510632, China
| | | | | | | |
Collapse
|
31
|
Badal SS, Danesh FR. New insights into molecular mechanisms of diabetic kidney disease. Am J Kidney Dis 2014; 63:S63-83. [PMID: 24461730 DOI: 10.1053/j.ajkd.2013.10.047] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 10/08/2013] [Indexed: 01/12/2023]
Abstract
Diabetic kidney disease remains a major microvascular complication of diabetes and the most common cause of chronic kidney failure requiring dialysis in the United States. Medical advances over the past century have substantially improved the management of diabetes mellitus and thereby have increased patient survival. However, current standards of care reduce but do not eliminate the risk of diabetic kidney disease, and further studies are warranted to define new strategies for reducing the risk of diabetic kidney disease. In this review, we highlight some of the novel and established molecular mechanisms that contribute to the development of the disease and its outcomes. In particular, we discuss recent advances in our understanding of the molecular mechanisms implicated in the pathogenesis and progression of diabetic kidney disease, with special emphasis on the mitochondrial oxidative stress and microRNA targets. Additionally, candidate genes associated with susceptibility to diabetic kidney disease and alterations in various cytokines, chemokines, and growth factors are addressed briefly.
Collapse
Affiliation(s)
- Shawn S Badal
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX
| | - Farhad R Danesh
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX; Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
32
|
ALK1-Smad1/5 signaling pathway in fibrosis development: friend or foe? Cytokine Growth Factor Rev 2013; 24:523-37. [PMID: 24055043 DOI: 10.1016/j.cytogfr.2013.08.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 08/14/2013] [Indexed: 12/29/2022]
Abstract
Fibrosis is a common phenomenon associated with several pathologies, characterized by an excessive extracellular matrix deposition that leads to a progressive organ dysfunction. Thus fibrosis has a relevant role in chronic diseases affecting the kidney, the liver, lung, skin (scleroderma) and joints (arthritis), among others. The pathogenesis of fibrosis in different organs share numerous similarities, being one of them the presence of activated fibroblasts, denominated myofibroblast, which act as the main source of extracellular matrix proteins. Transforming growth factor beta-1 (TGF-β1) is a profibrotic cytokine that plays a pivotal role in fibrosis. The TGF-β1/ALK5/Smad3 signaling pathway has been studied in fibrosis extensively. However, an increasing number of studies involving the ALK1/Smad1 pathway in the fibrotic process exist. In this review we offer a perspective of the function of ALK1/Smad1 pathway in renal fibrosis, liver fibrosis, scleroderma and osteoarthritis, suggesting this pathway as a powerful therapeutical target. We also propose several strategies to modulate the activity of this pathway and its consequences in the fibrotic process.
Collapse
|
33
|
Gu L, Johno H, Nakajima S, Kato H, Takahashi S, Katoh R, Kitamura M. Blockade of Smad signaling by 3'-deoxyadenosine: a mechanism for its anti-fibrotic potential. J Transl Med 2013; 93:450-61. [PMID: 23439432 DOI: 10.1038/labinvest.2013.4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Cordyceps militaris has been used in Eastern countries for the treatment of various diseases including chronic kidney diseases. However, there are no reports that identified its active entities and molecular mechanisms underlying its therapeutic effectiveness. 3'-Deoxyadenosine is a major nucleoside derivative isolated from C. militaris. Some reports suggested that both C. militaris and 3'-deoxyadenosine have anti-inflammatory and anti-fibrotic effects. In the present report, we investigated whether and how 3'-deoxyadenosine interferes with fibrogenic processes in the kidney. For this purpose, we examined effects of 3'-deoxyadenosine on the expression of collagens triggered by transforming growth factor-β (TGF-β1) and bone morphogenetic protein-4 (BMP-4), especially focusing on the regulation of Smad signaling in vitro and in vivo. We found that 3'-deoxyadenosine suppressed expression of collagens induced by TGF-β1 and BMP-4 dose dependently. This suppression occurred at the transcriptional level and was correlated with blunted activation of the CAGA box and the BMP-responsive element. The suppressive effect on the TGF-β/BMP signaling was mediated mainly by adenosine transporter and partially by the A3 adenosine receptor, but not A1/A2 adenosine receptors. 3'-Deoxyadenosine reduced levels of both phosphorylated and total Smad proteins (Smad1, 2 and 3) dose dependently. It was mainly ascribed to transcriptional suppression, but not to enhanced protein degradation and eIF2α-mediated translational suppression. Consistent with the in vitro results, in vivo administration with 3'-deoxyadenosine reduced the levels of phosphorylated and total Smad proteins, as well as the levels of Smad mRNAs, in the kidney subjected to unilateral ureteral obstruction. It was associated with blunted induction of type I collagen and α-smooth muscle actin, a decrease in the number of interstitial myofibroblasts and reduced fibrotic area. These results suggest that 3'-deoxyadenosine interferes with the TGF-β and BMP signaling via downregulation of Smads, which may underlie the anti-fibrotic effect of this agent. 3'-Deoxyadenosine may be useful for therapeutic intervention in various TGF-β-related fibrotic disorders.
Collapse
Affiliation(s)
- Liubao Gu
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Torikoshi K, Abe H, Matsubara T, Hirano T, Ohshima T, Murakami T, Araki M, Mima A, Iehara N, Fukatsu A, Kita T, Arai H, Doi T. Protein inhibitor of activated STAT, PIASy regulates α-smooth muscle actin expression by interacting with E12 in mesangial cells. PLoS One 2012; 7:e41186. [PMID: 22829926 PMCID: PMC3400623 DOI: 10.1371/journal.pone.0041186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 06/18/2012] [Indexed: 01/05/2023] Open
Abstract
Phenotypic transformation of mesangial cells (MCs) is implicated in the development of glomerular disease; however, the mechanisms underlying their altered genetic program is still unclear. α-smooth muscle actin (α-SMA) is known to be a crucial marker for phenotypic transformation of MCs. Recently, E-boxes and the class I basic helix-loop-helix proteins, such as E12 have been shown to regulateα-SMA expression. Therefore, we tried to identify a novel E12 binding protein in MCs and to examine its role in glomerulonephritis. We found that PIASy, one of the protein inhibitors of activated STAT family protein, interacted with E12 by yeast two-hybrid screens and coimmunopreciptation assays. Overexpression of E12 significantly enhanced theα-SMA promoter activity, and the increase was blocked by co-transfection of PIASy, but not by a PIASy RING mutant. In vivo sumoylation assays revealed that PIASy was a SUMO E3 ligase for E12. Furthermore, transforming growth factor-β (TGF-β) treatment induced expression of both PIASy and E12, consistent with α-SMA expression. Moreover, reduced expression of PIASy protein by siRNA specific for PIASy resulted in increased TGF-β-mediated α-SMA expression. In vivo, PIASy and E12 were dramatically upregulated along with α-SMA and TGF-β in the proliferative phase of Thy1 glomerulonephritis. Furthermore, an association between PIASy and E12 proteins was observed at day 6 by IP-western blotting, but not at day 0. These results suggest that TGF-β up-regulates PIASy expression in MCs to down-regulateα-SMA gene transcription by the interaction with E12.
Collapse
Affiliation(s)
- Kazuo Torikoshi
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideharu Abe
- Department of Nephrology, Health-Bioscience Institute, University of Tokushima Graduate School of Medicine, Tokushima, Japan
| | - Takeshi Matsubara
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- * E-mail:
| | - Takahiro Hirano
- Department of Nephrology, Health-Bioscience Institute, University of Tokushima Graduate School of Medicine, Tokushima, Japan
| | - Takayuki Ohshima
- Faculty of Pharmaceutical Science at Kagawa Campus, Tokushima Bunri University, Kagawa, Japan
| | - Taichi Murakami
- Department of Nephrology, Health-Bioscience Institute, University of Tokushima Graduate School of Medicine, Tokushima, Japan
| | - Makoto Araki
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akira Mima
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriyuki Iehara
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Atsushi Fukatsu
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toru Kita
- Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hidenori Arai
- Department of Human Health Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshio Doi
- Department of Nephrology, Health-Bioscience Institute, University of Tokushima Graduate School of Medicine, Tokushima, Japan
| |
Collapse
|
35
|
Mechanical stretch down-regulates expression of the Smad6 gene in cultured rat mesangial cells. Clin Exp Nephrol 2012; 16:690-6. [DOI: 10.1007/s10157-012-0630-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 03/15/2012] [Indexed: 02/04/2023]
|
36
|
Abe H, Tominaga T, Matsubara T, Abe N, Kishi S, Nagai K, Murakami T, Araoka T, Doi T. Scleraxis modulates bone morphogenetic protein 4 (BMP4)-Smad1 protein-smooth muscle α-actin (SMA) signal transduction in diabetic nephropathy. J Biol Chem 2012; 287:20430-42. [PMID: 22474292 DOI: 10.1074/jbc.m111.275610] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Activation of mesangial cells (MCs), which is characterized by induction of smooth muscle α-actin (SMA) expression, contributes to a key event in various renal diseases; however, the mechanisms controlling MC differentiation are still largely undefined. Activated Smad1 induced SMA in a dose-dependent manner in MCs. As a direct regulating molecule for SMA, we identified and characterized scleraxis (Scx) as a new phenotype modulator in advanced glycation end product (AGE)-exposed MCs. Scx physically associated with E12 and bound the E-box in the promoter of SMA and negatively regulated the AGE-induced SMA expression. Scx induced expression and secretion of bone morphogenetic protein 4 (BMP4), thereby controlling the Smad1 activation in AGE-treated MCs. In diabetic mice, Scx was concomitantly expressed with SMA in the glomeruli. Inhibitor of differentiation 1 (Id1) was further induced by extended treatment with AGE, thereby dislodging Scx from the SMA promoter. These data suggest that Scx and Id1 are involved in the BMP4-Smad1-SMA signal transduction pathway besides the TGFβ1-Smad1-SMA signaling pathway and modulate phenotypic changes in MCs in diabetic nephropathy.
Collapse
Affiliation(s)
- Hideharu Abe
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School of Medicine, Tokushima 770-8503, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Ogawa S, Matsushima M, Okamura M, Senda M, Sakamoto T, Nako K, Ito S. Urinary type IV collagen excretion predicts an increased urinary albumin-to-creatinine ratio in normoalbuminuric patients with diabetes. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/jdm.2012.24065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Kishi S, Abe H, Akiyama H, Tominaga T, Murakami T, Mima A, Nagai K, Kishi F, Matsuura M, Matsubara T, Iehara N, Ueda O, Fukushima N, Jishage KI, Doi T. SOX9 protein induces a chondrogenic phenotype of mesangial cells and contributes to advanced diabetic nephropathy. J Biol Chem 2011; 286:32162-9. [PMID: 21795715 DOI: 10.1074/jbc.m111.244541] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Diabetic nephropathy (DN) is the most important chronic kidney disease. We previously reported that Smad1 transcriptionally regulates the expression of extracellular matrix in DN. Phenotypic change in mesangial cells (MCs) is a key pathologic event in the progression of DN. The aim of this study is to investigate a novel mechanism underlying chondrogenic phenotypic change in MCs that results in the development of DN. MCs showed chondrogenic potential in a micromass culture, and BMP4 induced the expression of chondrocyte markers (SRY-related HMG Box 9 (SOX9) and type II collagen (COL2)). Advanced glycation end products induced the expression of chondrocyte marker proteins downstream from the BMP4-Smad1 signaling pathway in MCs. In addition, hypoxia also induced the expression of BMP4, hypoxia-inducible factor-1α (HIF-1α), and chondrocyte markers. Overexpression of SOX9 caused ectopic expression of proteoglycans and COL2 in MCs. Furthermore, forced expression of Smad1 induced chondrocyte markers as well. Dorsomorphin inhibited these inductions. Glomerular expressions of HIF-1α, BMP4, and chondrocyte markers were observed in diabetic nephropathy mice. These positive stainings were observed in mesangial sclerotic lesions. SOX9 was partially colocalized with HIF-1α and BMP4 in diabetic glomeruli. BMP4 knock-in transgenic mice showed not only similar pathological lesions to DN, but also the induction of chondrocyte markers in the sclerotic lesions. Here we demonstrate that HIF-1α and BMP4 induce SOX9 expression and subsequent chondrogenic phenotype change in DN. The results suggested that the transdifferentiation of MCs into chondrocyte-like cells in chronic hypoxic stress may result in irreversible structural change in DN.
Collapse
Affiliation(s)
- Seiji Kishi
- Department of Nephrology, Graduate School of Medicine, Health-Bioscience Institute, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang Y, Wang DH. Protective effect of TRPV1 against renal fibrosis via inhibition of TGF-β/Smad signaling in DOCA-salt hypertension. Mol Med 2011; 17:1204-12. [PMID: 21792478 DOI: 10.2119/molmed.2011.00063] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 07/21/2011] [Indexed: 12/11/2022] Open
Abstract
To investigate the effects of the transient receptor potential vanilloid type 1 (TRPV1) channel on renal extracellular matrix (ECM) protein expression including collagen deposition and the transforming growth factor β (TGF-β)/Smad signaling pathway during salt-dependent hypertension, wild-type (WT) and TRPV1-null (TRPV1⁻/⁻) mutant mice were uninephrectomized and given deoxycorticosterone acetate (DOCA)-salt for 4 wks. TRPV1 gene ablation exaggerated DOCA-salt-induced impairment of renal function as evidenced by increased albumin excretion (μg/24 h) compared with WT mice (83.7 ± 7.1 versus 28.3 ± 4.8, P < 0.05), but had no apparent effect on mean arterial pressure (mmHg) as determined by radiotelemetry (141 ± 4 versus 138 ± 3, P > 0.05). Morphological analysis showed that DOCA-salt-induced glomerulosclerosis, tubular injury and macrophage infiltration (cells/mm²) were increased in TRPV1⁻/⁻ compared with WT mice (0.74 ± 0.08 versus 0.34 ± 0.04; 3.14 ± 0.26 versus 2.00 ± 0.31; 68 ± 5 versus 40 ± 4, P < 0.05). Immunostaining studies showed that DOCA-salt treatment decreased nephrin but increased collagen type I and IV as well as phosphorylated Smad2/3 staining in kidneys of TRPV1⁻/⁻ compared with WT mice. Hydroxyproline assay and Western blot showed that DOCA-salt treatment increased collagen content (μg/mg dry tissue) and fibronectin protein expression (%β-actin arbitrary units) in the kidney of TRPV1⁻/⁻ compared with WT mice (26.7 ± 2.7 versus 17.4 ± 1.8; 0.93 ± 0.07 versus 0.65 ± 0.08, P < 0.05). Acceleration of renal ECM protein deposition in DOCA-salt-treated TRPV1⁻/⁻ mice was accompanied by increased TGF-β1, as well as phosphorylation of Smad2/3 protein expression (%β-actin arbitrary units) compared with DOCA-salt-treated WT mice (0.61 ± 0.07 versus 0.32 ± 0.05; 0.57 ± 0.07 versus 0.25 ± 0.05; 0.71 ± 0.08 versus 0.40 ± 0.06, P < 0.05). These results show that exaggerated renal functional and structural injuries are accompanied by increased production of ECM protein and activation of the TGF-β/Smad2/3 signaling pathway. These data suggest that activation of TRPV1 attenuates the progression of renal fibrosis possibly via suppression of the TGF-β and its downstream regulatory signaling pathway.
Collapse
Affiliation(s)
- Youping Wang
- Central Laboratory for Basic Research in Medicine, and Division of Cardiology, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, China
| | | |
Collapse
|
40
|
CCN2 is required for the TGF-β induced activation of Smad1-Erk1/2 signaling network. PLoS One 2011; 6:e21911. [PMID: 21760921 PMCID: PMC3132735 DOI: 10.1371/journal.pone.0021911] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 06/14/2011] [Indexed: 11/19/2022] Open
Abstract
Connective tissue growth factor (CCN2) is a multifunctional matricellular protein, which is frequently overexpressed during organ fibrosis. CCN2 is a mediator of the pro-fibrotic effects of TGF-β in cultured cells, but the specific function of CCN2 in the fibrotic process has not been elucidated. In this study we characterized the CCN2-dependent signaling pathways that are required for the TGF-β induced fibrogenic response. By depleting endogenous CCN2 we show that CCN2 is indispensable for the TGF-β-induced phosphorylation of Smad1 and Erk1/2, but it is unnecessary for the activation of Smad3. TGF-β stimulation triggered formation of the CCN2/β3 integrin protein complexes and activation of Src signaling. Furthermore, we demonstrated that signaling through the αvβ3 integrin receptor and Src was required for the TGF-β induced Smad1 phosphorylation. Recombinant CCN2 activated Src and Erk1/2 signaling, and induced phosphorylation of Fli1, but was unable to stimulate Smad1 or Smad3 phosphorylation. Additional experiments were performed to investigate the role of CCN2 in collagen production. Consistent with the previous studies, blockade of CCN2 abrogated TGF-β-induced collagen mRNA and protein levels. Recombinant CCN2 potently stimulated collagen mRNA levels and upregulated activity of the COL1A2 promoter, however CCN2 was a weak inducer of collagen protein levels. CCN2 stimulation of collagen was dose-dependent with the lower doses (<50 ng/ml) having a stimulatory effect and higher doses having an inhibitory effect on collagen gene expression. In conclusion, our study defines a novel CCN2/αvβ3 integrin/Src/Smad1 axis that contributes to the pro-fibrotic TGF-β signaling and suggests that blockade of this pathway may be beneficial for the treatment of fibrosis.
Collapse
|
41
|
Tominaga T, Abe H, Ueda O, Goto C, Nakahara K, Murakami T, Matsubara T, Mima A, Nagai K, Araoka T, Kishi S, Fukushima N, Jishage KI, Doi T. Activation of bone morphogenetic protein 4 signaling leads to glomerulosclerosis that mimics diabetic nephropathy. J Biol Chem 2011; 286:20109-16. [PMID: 21471216 DOI: 10.1074/jbc.m110.179382] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Diabetic nephropathy (DN) is the most common cause of chronic kidney disease. We have previously reported that Smad1 transcriptionally regulates the expression of extracellular matrix (ECM) proteins in DN. However, little is known about the regulatory mechanisms that induce and activate Smad1. Here, bone morphogenetic protein 4 (Bmp4) was found to up-regulate the expression of Smad1 in mesangial cells and subsequently to phosphorylate Smad1 downstream of the advanced glycation end product-receptor for advanced glycation end product signaling pathway. Moreover, Bmp4 utilized Alk3 and affected the activation of Smad1 and Col4 expressions in mesangial cells. In the diabetic mouse, Bmp4 was remarkably activated in the glomeruli, and the mesangial area was expanded. To elucidate the direct function of Bmp4 action in the kidneys, we generated transgenic mice inducible for the expression of Bmp4. Tamoxifen treatment dramatically induced the expression of Bmp4, especially in the glomeruli of the mice. Notably, in the nondiabetic condition, the mice exhibited not only an expansion of the mesangial area and thickening of the basement membrane but also remarkable albuminuria, which are consistent with the distinct glomerular injuries in DN. ECM protein overexpression and activation of Smad1 in the glomeruli were also observed in the mice. The mesangial expansion in the mice was significantly correlated with albuminuria. Furthermore, the heterozygous Bmp4 knock-out mice inhibited the glomerular injuries compared with wild type mice in diabetic conditions. Here, we show that BMP4 may act as an upstream regulatory molecule for the process of ECM accumulation in DN and thereby reveals a new aspect of the molecular mechanisms involved in DN.
Collapse
Affiliation(s)
- Tatsuya Tominaga
- Department of Nephrology, Graduate School of Medicine, Health-Bioscience Institute, University of Tokushima, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mima A, Abe H, Nagai K, Arai H, Matsubara T, Araki M, Torikoshi K, Tominaga T, Iehara N, Fukatsu A, Kita T, Doi T. Activation of Src mediates PDGF-induced Smad1 phosphorylation and contributes to the progression of glomerulosclerosis in glomerulonephritis. PLoS One 2011; 6:e17929. [PMID: 21445358 PMCID: PMC3062564 DOI: 10.1371/journal.pone.0017929] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 02/20/2011] [Indexed: 01/03/2023] Open
Abstract
Platelet-derived growth factor (PDGF) plays critical roles in mesangial cell (MC) proliferation in mesangial proliferative glomerulonephritis. We showed previously that Smad1 contributes to PDGF-dependent proliferation of MCs, but the mechanism by which Smad1 is activated by PDGF is not precisely known. Here we examined the role of c-Src tyrosine kinase in the proliferative change of MCs. Experimental mesangial proliferative glomerulonephritis (Thy1 GN) was induced by a single intravenous injection of anti-rat Thy-1.1 monoclonal antibody. In Thy1 GN, MC proliferation and type IV collagen (Col4) expression peaked on day 6. Immunohistochemical staining for the expression of phospho-Src (pSrc), phospho-Smad1 (pSmad1), Col4, and smooth muscle α-actin (SMA) revealed that the activation of c-Src and Smad1 signals in glomeruli peaked on day 6, consistent with the peak of mesangial proliferation. When treated with PP2, a Src inhibitor, both mesangial proliferation and sclerosis were significantly reduced. PP2 administration also significantly reduced pSmad1, Col4, and SMA expression. PDGF induced Col4 synthesis in association with increased expression of pSrc and pSmad1 in cultured MCs. In addition, PP2 reduced Col4 synthesis along with decreased pSrc and pSmad1 protein expression in vitro. Moreover, the addition of siRNA against c-Src significantly reduced the phosphorylation of Smad1 and the overproduction of Col4. These results provide new evidence that the activation of Src/Smad1 signaling pathway plays a key role in the development of glomerulosclerosis in experimental glomerulonephritis.
Collapse
Affiliation(s)
- Akira Mima
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideharu Abe
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
- * E-mail:
| | - Kojiro Nagai
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Hidenori Arai
- Department of Geriatric Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Matsubara
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Makoto Araki
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuo Torikoshi
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Noriyuki Iehara
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Atsushi Fukatsu
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toru Kita
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshio Doi
- Department of Nephrology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| |
Collapse
|
43
|
Papachroni KK, Piperi C, Levidou G, Korkolopoulou P, Pawelczyk L, Diamanti-Kandarakis E, Papavassiliou AG. Lysyl oxidase interacts with AGE signalling to modulate collagen synthesis in polycystic ovarian tissue. J Cell Mol Med 2011; 14:2460-9. [PMID: 19583806 PMCID: PMC3823163 DOI: 10.1111/j.1582-4934.2009.00841.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Connective tissue components – collagen types I, III and IV – surrounding the ovarian follicles undergo drastic changes during ovulation. Abnormal collagen synthesis and increased volume and density of ovarian stroma characterize the polycystic ovary syndrome (PCOS). During the ovulatory process, collagen synthesis is regulated by prolyl hydroxylase and lysyl oxidase (LOX) activity in ovarian follicles. LOX catalyzes collagen and elastin cross-linking and plays essential role in coordinating the control of ovarian extracellular matrix (ECM) during follicular development. We have recently shown accumulation of advanced glycation end products (AGEs), molecules that stimulate ECM production and abnormal collagen cross-linking, in ovarian tissue. However, the possible link between LOX and AGEs-induced signalling in collagen production and stroma formation in ovarian tissue from PCOS remains elusive. The present study investigates the hypothesis of AGE signalling pathway interaction with LOX gene activity in polycystic ovarian (PCO) tissue. We show an increased distribution and co-localization of LOX, collagen type IV and AGE molecules in the PCO tissue compared to control, as well as augmented expression of AGE signalling mediators/effectors, phospho(p)-ERK, phospho(p)-c-Jun and nuclear factor κB (NF-κB) in pathological tissue. Moreover, we demonstrate binding of AGE-induced transcription factors, NF-κB and activator protein-1 (AP-1) on LOX promoter, indicating a possible involvement of AGEs in LOX gene regulation, which may account for the documented increase in LOX mRNA and protein levels compared to control. These findings suggest that deposition of excess collagen in PCO tissue that induces cystogenesis may, in part, be due to AGE-mediated stimulation of LOX activity.
Collapse
Affiliation(s)
- Katerina K Papachroni
- Department of Biological Chemistry, Medical School, University of Athens, Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
44
|
Araoka T, Abe H, Tominaga T, Mima A, Matsubara T, Murakami T, Kishi S, Nagai K, Doi T. Transcription factor 7-like 2 (TCF7L2) regulates activin receptor-like kinase 1 (ALK1)/Smad1 pathway for development of diabetic nephropathy. Mol Cells 2010; 30:209-18. [PMID: 20803090 DOI: 10.1007/s10059-010-0109-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 05/17/2010] [Accepted: 05/28/2010] [Indexed: 11/29/2022] Open
Abstract
Smad1 has previously been shown to play a key role in the development of diabetic nephropathy (DN), by increasing synthesis of extracellular matrix. However, the regulatory mechanism of Smad1 in DN is still unclear. This study aims to elucidate molecular interactions between activin receptor-like kinase 1 (ALK1)/Smad1 signaling pathway and transcription factor 7-like 2 (TCF7L2) in the progression of DN in vitro and in vivo. The expressions of TCF7L2 and ALK1 were induced by advanced glycation end products (AGEs) in parallel with Smad1, phosphorylated Smad1 (pSmad1), and alpha-smooth muscle actin (α-SMA) through TGF-β1 in cultured mesangial cells. Constitutively active ALK1 increased pSmad1 and α-SMA expressions. The binding of TCF7L2 to ALK1 promoter was confirmed by chromatin immunoprecipitation assay. Furthermore, TCF7L2 induced promoter activity of ALK1. AGEs and TGF-β1 induced a marked increase in TCF7L2 expression in parallel with ALK1. Overexpression of TCF7L2 increased the expressions of ALK1 and Smad1. Inversely, TCF7L2 knockdown by siRNA suppressed α-SMA expression as well as ALK1 and Smad1. The iNOS transgenic mice (iNOS-Tgm), which developed diabetic glomerulosclerosis resembling human diabetic nephropathy, exhibited markedly increased expressions of ALK1, TCF7L2, Smad1, pSmad1, and α-SMA in glomeruli in association with mesangial matrix expansion. These results provide a new evidence that the TCF7L2/ALK1/Smad1 pathway plays a key role in the development of DN.
Collapse
Affiliation(s)
- Toshikazu Araoka
- Deapartment of Nephrology, Graduate School of Medicine, Institute of Health-Bio-Science, University of Tokushima, Tokushima, 770-8503, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Persistent transforming growth factor beta (TGF-beta) signaling is the major factor contributing to scleroderma (SSc) fibrosis. This review will summarize recent progress on the noncanonical TGF-beta signaling pathways and their role in SSc fibrosis. RECENT FINDINGS Canonical TGF-beta signaling involves activation of the TGF-beta receptors and downstream signal transducers Smad2/3. The term noncanonical TGF-beta signaling includes a variety of intracellular signaling pathways activated by TGF-beta independently of Smad2/3 activation. There is evidence that these pathways play important role in SSc fibrosis. In a subset of SSc fibroblasts, a multiligand receptor complex consisting of TGF-beta and CCN2 receptors drives constitutive activation of the Smad1 pathway. CCN2 is also a primary effector of this pathway, thus establishing an autocrine loop that amplifies TGF-beta signaling. SSc fibroblasts also demonstrate reduced expression of endogenous antagonists of TGF-beta signaling including transcriptional repressors, Friend leukemia integration-1 and perixosome proliferator-activated receptor-gamma, as well as inhibitor of Smad3 phosphorylation, PTEN. PTEN is a key mediator of the cross-talk between the sphingosine kinase and the TGF-beta pathways. SUMMARY Discovery of the role of noncanonical TGF-beta signaling in fibrosis offers new molecular targets for the antifibrotic therapies. Due to the heterogeneous nature of SSc, knowledge of these pathways could help to tailor the therapy to the individual patient depending on the activation status of a specific profibrotic pathway.
Collapse
|
46
|
Wu J, Mei C, Vlassara H, Striker GE, Zheng F. Oxidative stress-induced JNK activation contributes to proinflammatory phenotype of aging diabetic mesangial cells. Am J Physiol Renal Physiol 2009; 297:F1622-31. [PMID: 19776174 DOI: 10.1152/ajprenal.00078.2009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chronic inflammation and increased oxidative stress (OS) play an important role in diabetic nephropathy progression. Herein, we show that mesangial cells from streptozotocin-induced aging diabetic mice, a model of progressive diabetic nephropathy, exhibited increased OS and a proinflammatory phenotype characterized by elevated chemokines and ICAM-1 expression. This phenotypic change was consistent with the extensive inflammatory lesions present in aging diabetic kidneys and was not found in mesangial cells from old and young controls or young diabetic mice. Activation of the c-Jun NH(2)-terminal kinase (JNK) pathway was a likely contributor to the proinflammatory phenotype of aging diabetic mesangial cells since 1) phosphorylated JNK levels and JNK kinase activity were increased in these cells, 2) suppression of JNK significantly decreased monocyte chemoattractant protein-1 (MCP-1) production in these cells, and 3) activation of JNK in normal mesangial cells induced inflammation. Elevated OS in aging diabetic mesangial cells may be a cause of JNK activation and inflammation, because antioxidant treatment decreased JNK phosphorylation and MCP-1 production. Additionally, decreased expression of mitogen-activated protein kinase phosphatase 5 (MKP5) may also contribute to increased JNK and inflammation in aging diabetic mesangial cells since overexpression of MKP5 in these cells normalized phosphorylated JNK levels and reversed the proinflammatory phenotype. Moreover, knocking down of MKP5 expression in old control mesangial cells resulted in JNK activation and MCP-1 production, a phenotype seen in aging diabetic mesangial cells. Interestingly, MKP5 phosphatase activity was diminished by free radicals in vitro. Thus, OS may induce inflammation in mesangial cells by activating JNK through either a direct activation of JNK or indirectly by suppression of MKP5 activity. Proinflammatory phenotype of mesangial cells may contribute to chronic inflammatory lesions and disease progression of aging diabetic mice.
Collapse
Affiliation(s)
- Jin Wu
- Divison of Experimental Diabetes and Aging, Department of Geriatrics, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | |
Collapse
|
47
|
McKnight AJ, Woodman AM, Parkkonen M, Patterson CC, Savage DA, Forsblom C, Pettigrew KA, Sadlier D, Groop PH, Maxwell AP. Investigation of DNA polymorphisms in SMAD genes for genetic predisposition to diabetic nephropathy in patients with type 1 diabetes mellitus. Diabetologia 2009; 52:844-9. [PMID: 19247629 DOI: 10.1007/s00125-009-1281-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 01/13/2009] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS SMAD proteins are involved in multiple signalling pathways and are key modulators of gene expression. We hypothesised that genetic variation in selected SMAD genes contributes to susceptibility to diabetic nephropathy. METHODS We selected 13 haplotype tag (ht) single nucleotide polymorphisms (SNPs) from 67 variants identified by resequencing the SMAD2 and SMAD3 genes. For SMAD1, SMAD4 and SMAD5 genes, genotype data were downloaded for 217 SNPs from Phase II of the International HapMap project. Of these, 85 SNPs met our inclusion criteria, resulting in the selection of 13 tag SNPs for further investigation. A case-control approach was employed, using 267 nephropathic patients and 442 controls with type 1 diabetes from Ireland. Two further populations (totalling 1,407 patients, 2,238 controls) were genotyped to validate initial findings. Genotyping was conducted using iPLEX, TaqMan and gel electrophoresis. RESULTS The distribution of genotypes was in Hardy-Weinberg equilibrium. Analysis by the chi(2) test of genotype and allele frequencies in patients versus controls in the Irish population (n = 709) revealed evidence for the association of one allele at 5% level of significance (rs10515478, p(uncorrected) = 0.006; p(corrected) = 0.04). This finding represents a relatively small difference in allele frequency of 6.4% in the patient group compared with 10.7% in the control group; this difference was not supported in subsequent investigations using DNA from European individuals with similar phenotypic characteristics. CONCLUSIONS/INTERPRETATION We selected an appropriate subset of variants for the investigation of common genetic risk factors and assessed SMAD1 to SMAD5 genes for association with diabetic nephropathy. We conclude that common polymorphisms in these genes do not strongly influence genetic susceptibility to diabetic nephropathy in white individuals with type 1 diabetes mellitus.
Collapse
Affiliation(s)
- A J McKnight
- Nephrology Research Group, Queen's University of Belfast, c/o Regional Genetics Centre, Belfast City Hospital, Belfast, Northern Ireland, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Doi T, Mima A, Matsubara T, Tominaga T, Arai H, Abe H. The current clinical problems for early phase of diabetic nephropathy and approach for pathogenesis of diabetic nephropathy. Diabetes Res Clin Pract 2008; 82 Suppl 1:S21-4. [PMID: 18842318 DOI: 10.1016/j.diabres.2008.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The important clinical problems of diabetic nephropathy are both proteinuria and decrease of renal function. Pathological analysis showed decrease of GFR was correlated to degree of mesangial expansion but not thickening of GBM nor the other findings in human type 1 diabetic nephropathy. From the perspective in renal dysfunction, mesangial matrix expansion was crucial for diabetic nephropathy. However, there was no difference of mesangial expansion between normal and microalbuminuria stage in type 1 and 2 diabetes mellitus (DM). On the other hand, microalbuminuria definitely shows a key related factor for cardiovascular events, but it does not indicate a clear interaction for glomerulosclerosis. We need to search a new clinical marker for renal injury. We have first shown that Smad1 is a transcription factor for alpha1 and 2 of type 4 collagen (Col4), which is a major component of glomerulosclerosis. We have also identified Smad1 is a critical responsible molecule for developing glomerulosclerosis in rat diabetic nephropathy. We have found the good correlation between glomerulosclerosis and urinary Smad1 but not between glomerulosclerosis and urine albumin. These data suggests that urine Smad1 is a promising clinical marker for underlying glomerular damages in early stage diabetic nephropathy. The study also implicates that angiotensin II (AngII)-Src-Smad1 signaling pathway has played a key role for development of diabetic nephropathy. These suggest that it is necessary to clarify the whole mechanism related to Smad1 to identify the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Toshio Doi
- Department of Clinical Biology and Medicine, Graduate school of Medicine, Institute of Health Bio-Science, The University of Tokushima, Tokushima, Japan.
| | | | | | | | | | | |
Collapse
|
49
|
Type IV collagens regulate BMP signalling in Drosophila. Nature 2008; 455:72-7. [PMID: 18701888 DOI: 10.1038/nature07214] [Citation(s) in RCA: 257] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 06/26/2008] [Indexed: 12/14/2022]
Abstract
Dorsal-ventral patterning in vertebrate and invertebrate embryos is mediated by a conserved system of secreted proteins that establishes a bone morphogenetic protein (BMP) gradient. Although the Drosophila embryonic Decapentaplegic (Dpp) gradient has served as a model to understand how morphogen gradients are established, no role for the extracellular matrix has been previously described. Here we show that type IV collagen extracellular matrix proteins bind Dpp and regulate its signalling in both the Drosophila embryo and ovary. We provide evidence that the interaction between Dpp and type IV collagen augments Dpp signalling in the embryo by promoting gradient formation, yet it restricts the signalling range in the ovary through sequestration of the Dpp ligand. Together, these results identify a critical function of type IV collagens in modulating Dpp in the extracellular space during Drosophila development. On the basis of our findings that human type IV collagen binds BMP4, we predict that this role of type IV collagens will be conserved.
Collapse
|
50
|
Pannu J, Asano Y, Nakerakanti S, Smith E, Jablonska S, Blaszczyk M, ten Dijke P, Trojanowska M. Smad1 pathway is activated in systemic sclerosis fibroblasts and is targeted by imatinib mesylate. ACTA ACUST UNITED AC 2008; 58:2528-37. [DOI: 10.1002/art.23698] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|