1
|
Egal ESA, Kamdem SD, Yoshigi M, Yang CC, Pellizzari S, Kameni EM, Helms MN, Assassi S, Henkemeyer M, Frech TM, Mimche PN. EphB2 Receptor Promotes Dermal Fibrosis in Systemic Sclerosis. Arthritis Rheumatol 2024; 76:1303-1316. [PMID: 38589317 PMCID: PMC11288787 DOI: 10.1002/art.42858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 02/20/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
OBJECTIVE Erythropoietin-producing hepatocellular (Eph)/Ephrin cell-cell signaling is emerging as a key player in tissue fibrogenesis. The aim of this study was to test the hypothesis that the receptor tyrosine kinase EphB2 mediates dermal fibrosis in systemic sclerosis (SSc). METHODS We assessed normal and SSc human skin biopsies for EphB2 expression. The in vivo role of EphB2 in skin fibrosis was investigated by subjecting EphB2-knockout mice to both bleomycin-induced and tight skin (Tsk1/+) genetic mouse models of skin fibrosis. EphB2 kinase-dead and overactive point mutant mice were used to evaluate the role of EphB2 forward signaling in bleomycin-induced dermal fibrosis. In vitro studies were performed on dermal fibroblasts from patients with SSc and healthy controls, which was followed by in vivo analysis of fibroblast-specific Ephb2-deficient mice. RESULTS Expression of EphB2 is up-regulated in SSc skin tissue and explanted SSc dermal fibroblasts compared with healthy controls. EphB2 expression is elevated in two animal models of dermal fibrosis. In mice, EphB2 drives dermal fibrosis in both the bleomycin and the Tsk1/+ models of skin fibrosis. EphB2 forward signaling is a critical mediator of dermal fibrosis. Transforming growth factor-β (TGF-β) cytokines up-regulate EphB2 in dermal fibroblasts via noncanonical TGF-β/mother against decapentaplegic signaling, and silencing EPHB2 in human dermal fibroblasts is sufficient to dampen TGF-β-induced fibroblast-to-myofibroblast differentiation. Moreover, mice with fibroblast-specific deletion of EphB2 showed impaired fibroblast-to-myofibroblast differentiation and reduced skin fibrosis upon bleomycin challenge. CONCLUSION Our data implicate TGF-β regulation of EphB2 overexpression and kinase-mediated forward signaling in the development of dermal fibrosis in SSc. EphB2 thus represents a potential new therapeutic target for SSc.
Collapse
Affiliation(s)
- Erika SA Egal
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT84112, USA
| | - Severin Donald Kamdem
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT84112, USA
| | - Masaaki Yoshigi
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT84112, USA
| | - Ching-Chu Yang
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT84112, USA
| | - Sarah Pellizzari
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT84112, USA
| | - Ernest M Kameni
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT84112, USA
| | - My N Helms
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT84132, USA
| | - Shervin Assassi
- Division of Rheumatology, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX77030
| | - Mark Henkemeyer
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX75390, USA
| | - Tracy M Frech
- Vanderbilt University Medical Center, Division of Rheumatology and Immunology, Nashville, TN 37232, USA
| | - Patrice N Mimche
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT84112, USA
| |
Collapse
|
2
|
Valencia LJ, Tseng M, Chu ML, Yu L, Adedeji AO, Kiyota T. Zoledronic acid and ibandronate-induced nephrotoxicity in 2D and 3D proximal tubule cells derived from human and rat. Toxicol Sci 2024; 198:86-100. [PMID: 38059598 DOI: 10.1093/toxsci/kfad123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Drug-induced proximal tubule (PT) injury remains a serious safety concern throughout drug development. Traditional in vitro 2-dimensional (2D) and preclinical in vivo models often fail to predict drug-related injuries presented in clinical trials. Various 3-dimensional (3D) microphysiological systems (MPSs) have been developed to mimic physiologically relevant properties, enabling them to be more predictive toward nephrotoxicity. To explore the capabilities of an MPS across species, we compared cytotoxicity in hRPTEC/TERT1s and rat primary proximal tubular epithelial cells (rPPTECs) following exposure to zoledronic acid and ibandronate (62.5-500 µM), and antibiotic polymyxin B (PMB) (50 and 250 µM, respectively). For comparison, we investigated cytotoxicity using 2D cultured hRPTEC/TERT1s and rPPTECs following exposure to the same drugs, including overlapping concentrations, as their 3D counterparts. Regardless of the in vitro model, bisphosphonate-exposed rPPTECs exhibited cytotoxicity quicker than hRPTEC/TERT1s. PMB was less sensitive toward nephrotoxicity in rPPTECs than hRPTEC/TERT1s, demonstrating differences in species sensitivity within both 3D and 2D models. Generally, 2D cultured cells experienced faster drug-induced cytotoxicity compared to the MPSs, suggesting that MPSs can be advantageous for longer-term drug-exposure studies, if warranted. Furthermore, ibandronate-exposed hRPTEC/TERT1s and rPPTECs produced higher levels of inflammatory and kidney injury biomarkers compared to zoledronic acid, indicating that ibandronate induces acute kidney injury, but also a potential protective response since ibandronate is less toxic than zoledronic acid. Our study suggests that the MPS model can be used for preclinical screening of compounds prior to animal studies and human clinical trials.
Collapse
Affiliation(s)
- Leslie J Valencia
- Investigative Toxicology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
- Pathology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Min Tseng
- Investigative Toxicology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Mei-Lan Chu
- Pathology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Lanlan Yu
- Investigative Toxicology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Adeyemi O Adedeji
- Pathology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| | - Tomomi Kiyota
- Investigative Toxicology, Department of Safety Assessment, Genentech Inc., South San Francisco, California 94080, USA
| |
Collapse
|
3
|
Wang X, Richard ML, Caldwell TS, Sundararaj K, Sato S, Nowling TK, Zhang XK. Role of the transcription factor Fli-1 on the CXCL10/CXCR3 Axis. Front Immunol 2023; 14:1219279. [PMID: 37790939 PMCID: PMC10543418 DOI: 10.3389/fimmu.2023.1219279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/29/2023] [Indexed: 10/05/2023] Open
Abstract
The transcription factor Fli-1, a member of the ETS family of transcription factors, is implicated in the pathogenesis of lupus disease. Reduced Fli-1 expression in lupus mice leads to decreased renal Cxcl10 mRNA levels and renal infiltrating CXCR3+ T cells that parallels reduced renal inflammatory cell infiltration and renal damage. Inflammatory chemokine CXCL10 is critical for attracting inflammatory cells expressing the chemokine receptor CXCR3. The CXCL10/CXCR3 axis plays a role in the pathogenesis of various inflammatory diseases including lupus. Our data here demonstrate that renal CXCL10 protein levels are significantly lower in Fli-1 heterozygous MRL/lpr mice compared to wild-type MRL/lpr mice. Knockdown of Fli-1 significantly reduced CXCL10 secretion in mouse and human endothelial cells, and human mesangial cells, upon LPS or TNFα stimulation. The Fli-1 inhibitor, Camptothecin, significantly reduced CXCL10 production in human monocyte cells upon interferon stimulation. Four putative Ets binding sites in the Cxcl10 promoter showed significant enrichment for FLI-1; however, FLI-1 did not directly drive transcription from the human or mouse promoters, suggesting FLI-1 may regulate CXCL10 expression indirectly. Our results also suggest that the DNA binding domain of FLI-1 is necessary for regulation of human hCXCR3 promotor activity in human T cells and interactions with co-activators. Together, these results support a role for FLI-1 in modulating the CXCL10-CXCR3 axis by directly or indirectly regulating the expression of both genes to impact lupus disease development. Signaling pathways or drugs that reduce FLI-1 expression may offer novel approaches to lupus treatment.
Collapse
Affiliation(s)
- Xuan Wang
- Department of General Practice, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Mara Lennard Richard
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Tomika S. Caldwell
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Kamala Sundararaj
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Shuzo Sato
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tamara K. Nowling
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Xian K. Zhang
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
4
|
Wells A. Role of CXCR3 in fibrotic tissue responses. Int J Biochem Cell Biol 2022; 152:106311. [PMID: 36195287 DOI: 10.1016/j.biocel.2022.106311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022]
Abstract
Development of fibrosis leads to end stage diseases that defy treatments across all organs. This ensues as chronic inflammation is not dampened by physiologic processes that issue in the resolution phase of wound healing. Thus, these conditions can be considered diseases of "failure to heal". In the absence of broadly viable treatments, it is proposed to examine key switches in wound healing resolution to seek insights into novel approaches. Signaling through the GPCR CXCR3 has been shown to be one such critical player in this physiologic transition that limits and even reverses early fibrosis. As such, a number of investigators and early stage technology companies have posited that triggering this signaling network would limit fibrosis. While there are some conflicting results, a consensus is emerging that pharmacologic interventions that promote signaling through this pathway represent innovative ways to limit fibrotic diseases.
Collapse
Affiliation(s)
- Alan Wells
- Departments of Pathology, Bioengineering, and Computational & Systems Biology, and McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; R&D Service, Pittsburgh VA Health System, Pittsburgh, PA 15213, USA.
| |
Collapse
|
5
|
do Nascimento WRC, Nóbrega CGDO, Fernandes EDS, Santos PDA, Melo FL, Albuquerque MCPDA, de Lorena VMB, Costa VMA, Barbosa CCGS, de Souza VMO. Schistosoma mansoni infection decreases IL-33-mRNA expression and increases CXCL9 and CXCL10 production by peripheral blood cells. Med Microbiol Immunol 2022; 211:211-218. [PMID: 35819523 DOI: 10.1007/s00430-022-00745-6] [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: 06/13/2021] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
Schistosoma mansoni infections, particularly egg antigens, induce Th2-dominant granulomatous responses accompanied by remarkable immunoregulatory mechanisms that avoid intense fibrosis. Interleukin (IL)-33 is a cytokine that stimulates the early activation of Th2 responses, and its soluble ST2 receptor (sST2) avoids granulomatous response, as well as CXCL9 and CXCL10 chemokines that have antifibrotic activity. However, in schistosomiasis, these molecules have not been suitably studied. Therefore, this study aimed to measure IL-33 and sST2 RNA, cytokines, and chemokines in peripheral blood cultures from individuals living in schistosomiasis-endemic areas. Peripheral blood cells from individuals with S. mansoni (n = 34) and non-infected individuals (n = 31) were cultured under mitogen stimulation. Supernatant chemokines and cytokines were evaluated using a cytometric bead array, and IL-33 and sST2 mRNA expression was measured using qPCR. Infected individuals showed higher levels of CXCL8, CXCL9, CXCL10, IFN-γ, TNF-α, IL-6, IL-2, IL-4, and IL-10; there was a lower expression of IL-33 mRNA and similar expression of sST2mRNA in infected than non-infected individuals. In conclusion, for the first time, we demonstrated lower IL-33mRNA expression and high levels of the antifibrotic chemokines CXCL9 and CXCL10 in schistosomiasis mansoni, which could control exacerbations of the disease in individuals from endemic areas.
Collapse
Affiliation(s)
| | - Cassia Giselle de Oliveira Nóbrega
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, s/n. Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Erica de Souza Fernandes
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, s/n. Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Patrícia d'Emery Alves Santos
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, s/n. Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Fábio Lopes Melo
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Brazil
| | - Mônica Camelo Pessôa de Azevedo Albuquerque
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco, Recife, Brazil.,Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, s/n. Cidade Universitária, Recife, PE, 50670-901, Brazil
| | | | - Vláudia Maria Assis Costa
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco, Recife, Brazil.,Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, s/n. Cidade Universitária, Recife, PE, 50670-901, Brazil
| | | | - Valdênia Maria Oliveira de Souza
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, s/n. Cidade Universitária, Recife, PE, 50670-901, Brazil. .,Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, s/n. Cidade Universitária, Recife, PE, 50670-901, Brazil.
| |
Collapse
|
6
|
Magkrioti C, Antonopoulou G, Fanidis D, Pliaka V, Sakellaropoulos T, Alexopoulos LG, Ullmer C, Aidinis V. Lysophosphatidic Acid Is a Proinflammatory Stimulus of Renal Tubular Epithelial Cells. Int J Mol Sci 2022; 23:ijms23137452. [PMID: 35806457 PMCID: PMC9267536 DOI: 10.3390/ijms23137452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic kidney disease (CKD) refers to a spectrum of diseases defined by renal fibrosis, permanent alterations in kidney structure, and low glomerular-filtration rate. Prolonged epithelial-tubular damage involves a series of changes that eventually lead to CKD, highlighting the importance of tubular epithelial cells in this process. Lysophosphatidic acid (LPA) is a bioactive lipid that signals mainly through its six cognate LPA receptors and is implicated in several chronic inflammatory pathological conditions. In this report, we have stimulated human proximal tubular epithelial cells (HKC-8) with LPA and 175 other possibly pathological stimuli, and simultaneously detected the levels of 27 intracellular phosphoproteins and 32 extracellular secreted molecules with multiplex ELISA. This quantification revealed a large amount of information concerning the signaling and the physiology of HKC-8 cells that can be extrapolated to other proximal tubular epithelial cells. LPA responses clustered with pro-inflammatory stimuli such as TNF and IL-1, promoting the phosphorylation of important inflammatory signaling hubs, including CREB1, ERK1, JUN, IκΒα, and MEK1, as well as the secretion of inflammatory factors of clinical relevance, including CCL2, CCL3, CXCL10, ICAM1, IL-6, and IL-8, most of them shown for the first time in proximal tubular epithelial cells. The identified LPA-induced signal-transduction pathways, which were pharmacologically validated, and the secretion of the inflammatory factors offer novel insights into the possible role of LPA in CKD pathogenesis.
Collapse
Affiliation(s)
- Christiana Magkrioti
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (C.M.); (G.A.); (D.F.)
| | - Georgia Antonopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (C.M.); (G.A.); (D.F.)
| | - Dionysios Fanidis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (C.M.); (G.A.); (D.F.)
| | - Vaia Pliaka
- ProtATonce Ltd., 15343 Athens, Greece; (V.P.); (T.S.); (L.G.A.)
| | | | - Leonidas G. Alexopoulos
- ProtATonce Ltd., 15343 Athens, Greece; (V.P.); (T.S.); (L.G.A.)
- School of Mechanical Engineering, National Technical University of Athens, 15780 Zografou, Greece
| | - Christoph Ullmer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland;
| | - Vassilis Aidinis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (C.M.); (G.A.); (D.F.)
- Correspondence:
| |
Collapse
|
7
|
Dou M, Ding C, Zheng B, Deng G, Zhu K, Xu C, Xue W, Ding X, Zheng J, Tian P. Immune-Related Genes for Predicting Future Kidney Graft Loss: A Study Based on GEO Database. Front Immunol 2022; 13:859693. [PMID: 35281025 PMCID: PMC8913884 DOI: 10.3389/fimmu.2022.859693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
Objective We aimed to identify feature immune-related genes that correlated with graft rejection and to develop a prognostic model based on immune-related genes in kidney transplantation. Methods Gene expression profiles were obtained from the GEO database. The GSE36059 dataset was used as a discovery cohort. Then, differential expression analysis and a machine learning method were performed to select feature immune-related genes. After that, univariate and multivariate Cox regression analyses were used to identify prognosis-related genes. A novel Riskscore model was built based on the results of multivariate regression. The levels of these feature genes were also confirmed in an independent single-cell dataset and other GEO datasets. Results 15 immune-related genes were expressed differently between non-rejection and rejection kidney allografts. Those differentially expressed immune-related genes (DE-IRGs) were mainly associated with immune-related biological processes and pathways. Subsequently, a 5-immune-gene signature was constructed and showed favorable predictive results in the GSE21374 dataset. Recipients were divided into the high-risk and low-risk groups according to the median value of RiskScore. The GO and KEGG analysis indicated that the differentially expressed genes (DEGs) between high-risk and low-risk groups were mainly involved in inflammatory pathways, chemokine-related pathways, and rejection-related pathways. Immune infiltration analysis demonstrated that RiskScore was potentially related to immune infiltration. Kaplan-Meier survival analysis suggested that recipients in the high-risk group had poor graft survival. AUC values of 1- and 3-year graft survival were 0.804 and 0.793, respectively. Conclusion Our data suggest that this immune-related prognostic model had good sensitivity and specificity in predicting the 1- and 3-year kidney graft survival and might act as a useful tool for predicting kidney graft loss.
Collapse
Affiliation(s)
- Meng Dou
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chenguang Ding
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bingxuan Zheng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ge Deng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Kun Zhu
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Cuixiang Xu
- Center of Shaanxi Provincial Clinical Laboratory, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Wujun Xue
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoming Ding
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jin Zheng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Puxun Tian
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Puxun Tian,
| |
Collapse
|
8
|
Gao J, Wu L, Zhao Y, Hong Q, Feng Z, Chen X. Cxcl10 deficiency attenuates renal interstitial fibrosis through regulating epithelial-to-mesenchymal transition. Exp Cell Res 2022; 410:112965. [PMID: 34896075 DOI: 10.1016/j.yexcr.2021.112965] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/03/2021] [Accepted: 12/04/2021] [Indexed: 11/28/2022]
Abstract
IFN-γ-inducible protein 10 (IP-10, CXCL10) has been widely demonstrated to be involved in multiple kidney pathological processes. However, the role of CXCL10 in renal fibrosis remains unclear. In this study, Cxcl10-deficient (Cxcl10-/-) mice were used to generate the unilateral ureteral obstruction (UUO) model. The level of renal fibrosis and inflammatory cell infiltration was examined in vivo and the effects of CXCL10 on EMT process of HK-2 cells was investigated in vitro. We observed that the injury degree of renal tissue and the collagen deposition levels were lighter and the expression of α-SMA, collagen I and fibronectin was significantly reduced in Cxcl10-/- mice, while the expression of E-cadherin was increased. However, interstitial F4/80-positive macrophages and CD4-positive T lymphocytes were unaffected by knockout of Cxcl10. Furthermore, IFN-γ or CXCL10 stimulation could obviously promote the expression of α-SMA, collagen I, fibronectin and reduce the expression of E-cadherin in HK-2 cells, which could be inhibited by transfection of Cxcl10-siRNA. Our findings suggested Cxcl10 knockout could reduce renal dysfunction and inhibit renal fibrosis through regulating EMT process of renal tubular epithelial cells in murine UUO model. These results may provide a novel insight into the mechanism and a potential therapy target of renal fibrosis.
Collapse
Affiliation(s)
- Jie Gao
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, China; Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jingwu Road 324, Jinan, 250021, China
| | - Lingling Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, China
| | - Yinghua Zhao
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, China
| | - Quan Hong
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, China
| | - Zhe Feng
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing, 100853, China.
| |
Collapse
|
9
|
Mikolajczyk TP, Szczepaniak P, Vidler F, Maffia P, Graham GJ, Guzik TJ. Role of inflammatory chemokines in hypertension. Pharmacol Ther 2020; 223:107799. [PMID: 33359600 DOI: 10.1016/j.pharmthera.2020.107799] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Hypertension is associated with immune cells activation and their migration into the kidney, vasculature, heart and brain. These inflammatory mechanisms are critical for blood pressure regulation and mediate target organ damage, creating unique novel targets for pharmacological modulation. In response to angiotensin II and other pro-hypertensive stimuli, the expression of several inflammatory chemokines and their receptors is increased in the target organs, mediating homing of immune cells. In this review, we summarize the contribution of key inflammatory chemokines and their receptors to increased accumulation of immune cells in target organs and effects on vascular dysfunction, remodeling, oxidative stress and fibrosis, all of which contribute to blood pressure elevation. In particular, the role of CCL2, CCL5, CXCL8, CXCL9, CXCL10, CXCL11, CXCL16, CXCL1, CX3CL1, XCL1 and their receptors in the context of hypertension is discussed. Recent studies have tested the efficacy of pharmacological or genetic targeting of chemokines and their receptors on the development of hypertension. Promising results indicate that some of these pathways may serve as future therapeutic targets to improve blood pressure control and prevent target organ consequences including kidney failure, heart failure, atherosclerosis or cognitive impairment.
Collapse
Affiliation(s)
- Tomasz P Mikolajczyk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Francesca Vidler
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK; BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gerard J Graham
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
10
|
Feng Y, Guo F, Mai H, Liu J, Xia Z, Zhu G, Zhang J, Ma L, Fu P. Pterostilbene, a Bioactive Component of Blueberries, Alleviates Renal Interstitial Fibrosis by Inhibiting Macrophage-Myofibroblast Transition. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1715-1729. [PMID: 33148003 DOI: 10.1142/s0192415x20500858] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pterostilbene (PTB) is a derivative of resveratrol present in grapes and blueberries. PTB is structurally similar to resveratrol, possessing properties such as being analgesic, anti-aging, antidiabetic, anti-inflammatory, anti-obesity, anti-oxidation, cholesterol-reductive, and neuroprotective. However, there have not been reports on the effect of PTB on macrophage-myofibroblast transition (MMT) induced fibrosis in kidney. In this study, we investigated the antifibrotic effects of PTB on the in vivo mouse unilateral ureteral obstruction (UUO) model and in vitro MMT cells. Kidneys subjected to UUO with PTB treatment were collected for the investigation of PTB mediating MMT derived renal interstitial fibrosis. We conducted kidney RNA-seq transcriptomes and TGF-[Formula: see text]1-induced bone marrow-derived macrophages assays to determine the mechanisms of PTB. We found that PTB treatment suppressed the interstitial fibrosis in UUO mice. PTB also attenuated the number of MMT cells in vivo and in vitro. The transcriptomic analysis showed that CXCL10 may play a central role in the process of PTB-treated renal fibrosis. The siRNA-mediated CXCL10 knockdown decreased the number of MMT cells in TGF-[Formula: see text]1-induced bone marrow-derived macrophages. Our results suggested that PTB attenuated renal interstitial fibrosis by mediating MMT by regulating transcriptional activity of CXCL10.
Collapse
Affiliation(s)
- Yanhuan Feng
- Division of Nephrology and National Clinical, Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Fan Guo
- Division of Nephrology and National Clinical, Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Hongxia Mai
- Division of Nephrology and National Clinical, Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, P. R. China.,Department of Nephrology, Lanzhou University Second Hospital, Lanzhou 730030, P. R. China
| | - Jing Liu
- Division of Nephrology and National Clinical, Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Zijing Xia
- Division of Nephrology and National Clinical, Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Guonian Zhu
- Core Facility of West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Jinhang Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Liang Ma
- Division of Nephrology and National Clinical, Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Ping Fu
- Division of Nephrology and National Clinical, Research Center for Geriatrics, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, P. R. China
| |
Collapse
|
11
|
Growth Differentiation Factor 15 Ameliorates Anti-Glomerular Basement Membrane Glomerulonephritis in Mice. Int J Mol Sci 2020; 21:ijms21196978. [PMID: 32977372 PMCID: PMC7583818 DOI: 10.3390/ijms21196978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/16/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is a member of the transforming growth factor-β (TGF-β) cytokine family and an inflammation-associated protein. Here, we investigated the role of GDF15 in murine anti-glomerular basement membrane (GBM) glomerulonephritis. Glomerulonephritis induction in mice induced systemic expression of GDF15. Moreover, we demonstrate the protective effects for GDF15, as GDF15-deficient mice exhibited increased proteinuria with an aggravated crescent formation and mesangial expansion in anti-GBM nephritis. Herein, GDF15 was required for the regulation of T-cell chemotactic chemokines in the kidney. In addition, we found the upregulation of the CXCR3 receptor in activated T-cells in GDF15-deficient mice. These data indicate that CXCL10/CXCR3-dependent-signaling promotes the infiltration of T cells into the organ during acute inflammation controlled by GDF15. Together, these results reveal a novel mechanism limiting the migration of lymphocytes to the site of inflammation during glomerulonephritis.
Collapse
|
12
|
Premeaux TA, Javandel S, Hosaka KRJ, Greene M, Therrien N, Allen IE, Corley MJ, Valcour VG, Ndhlovu LC. Associations Between Plasma Immunomodulatory and Inflammatory Mediators With VACS Index Scores Among Older HIV-Infected Adults on Antiretroviral Therapy. Front Immunol 2020; 11:1321. [PMID: 32695109 PMCID: PMC7338430 DOI: 10.3389/fimmu.2020.01321] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
The prevalence of age-related comorbidities is increased in people living with HIV, even in those well-controlled on combination antiretroviral therapy (ART). Persistent immune activation and inflammation may play pivotal roles in the pathogenesis; however, the burden of morbidities in the older HIV infected population may be exacerbated and driven by distinct mechanisms. In a cross sectional study of 45 HIV-infected participants 60 years or older, we examined the relationships between 14 immunomodulatory and inflammatory factors and the Veterans Aging Cohort Study (VACS) Index, a metric of multimorbidity and mortality comprised of age, CD4 count, hemoglobin, Fibrosis-4 [FIB-4], and estimated glomerular filtration rate [eGFR], by linear regression analysis. All participants were virally suppressed (<50 HIV RNA copies/mL), on ART, and primarily Caucasian (86.7%), and male (91.1%). Plasma levels of monocyte/macrophage-associated (neopterin, IP-10, sCD163, sCD14, and MCP-1) and glycan-binding immunomodulatory factors (galectin (Gal)-1, Gal-3, and Gal-9) were assessed, as well as inflammatory biomarkers previously linked to the VACS Index (i.e., CRP, cystatin C, TNF-α, TNFRI, IL-6, and D-dimer) for comparison. In regression analysis, higher VACS index scores were associated with higher levels of neopterin, cystatin C, TNFRI, and Gal-9 (all p < 0.05), potentially driven by correlations found with individual VACS components, including age, CD4 count, FIB-4, and eGFR. Gal-9, cystatin C, and TNFRI directly correlated with the extent of multimorbidity. Multiple correlations among markers were observed, suggesting an interplay of overlapping, but distinct, pathways. Collectively, in addition to cystatin C and TNFRI, both galectin-9 and neopterin, independently emerged as novel fluid markers of the VACS Index and burden of comorbidity and may further guide in understanding pathogenic mechanisms of age-related disorders in older HIV-infected individuals on suppressive ART.
Collapse
Affiliation(s)
- Thomas A Premeaux
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Shireen Javandel
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Kalei R J Hosaka
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Meredith Greene
- Division of Geriatric Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Nicholas Therrien
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Michael J Corley
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Victor G Valcour
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States.,Division of Geriatric Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Lishomwa C Ndhlovu
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States.,Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
13
|
Feng Y, Li M, Wang S, Cong W, Hu G, Song Y, Xiao H, Dong E, Zhang Y. Paired box 6 inhibits cardiac fibroblast differentiation. Biochem Biophys Res Commun 2020; 528:561-566. [PMID: 32505347 DOI: 10.1016/j.bbrc.2020.05.146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/13/2020] [Indexed: 01/09/2023]
Abstract
Cardiac fibroblast (CF) differentiation plays a crucial role in cardiac fibrosis, which is a specific manifestation distinguishing pathological cardiac hypertrophy from physiological hypertrophy. The DNA-binding activity of paired box 6 (Pax6) has been shown to be oppositely regulated in physiological and pathological hypertrophy; however, it remains unclear whether Pax6 is involved in CF differentiation during cardiac fibrosis. We found that Pax6 is expressed in the heart of and CFs isolated from adult mice. Moreover, angiotensin II (Ang II) induced the downregulation of Pax6 mRNA and protein expression in fibrotic heart tissue and cardiac myofibroblasts. Pax6 knockdown in CFs promoted the expression of the myofibroblast marker α-smooth muscle actin (α-SMA) and the synthesis of the extracellular matrix (ECM) proteins collagen I and fibronectin. Furthermore, we validated the ability of Pax6 to bind to the promoter regions of Cxcl10 and Il1r2 and the intronic region of Tgfb1. Pax6 knockdown in CFs decreased CXC chemokine 10 (CXCL10) and interleukin-1 receptor 2 (IL-1R2) expression and increased transforming growth factor β1 (TGFβ1) expression, mimicking the effects of Ang II. In conclusion, Pax6 exerts an inhibitory effect on CF differentiation and ECM synthesis by transcriptionally activating the expression of the anti-fibrotic factors CXCL10 and IL-1R2 and repressing the expression of the pro-fibrotic factor TGFβ1. Therefore, maintaining Pax6 expression in CFs is essential for preventing CF differentiation, and provides a new therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Yenan Feng
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Mingzhe Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Shuaixing Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Wenwen Cong
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, School of Medicine Shihezi University, Shihezi, 832000, China.
| | - Guomin Hu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Yao Song
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
14
|
Gao J, Wu L, Wang S, Chen X. Role of Chemokine (C-X-C Motif) Ligand 10 (CXCL10) in Renal Diseases. Mediators Inflamm 2020; 2020:6194864. [PMID: 32089645 PMCID: PMC7025113 DOI: 10.1155/2020/6194864] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/02/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022] Open
Abstract
Chemokine C-X-C ligand 10 (CXCL10), also known as interferon-γ-inducible protein 10 (IP-10), exerts biological function mainly through binding to its specific receptor, CXCR3. Studies have shown that renal resident mesangial cells, renal tubular epithelial cells, podocytes, endothelial cells, and infiltrating inflammatory cells express CXCL10 and CXCR3 under inflammatory conditions. In the last few years, strong experimental and clinical evidence has indicated that CXCL10 is involved in the development of renal diseases through the chemoattraction of inflammatory cells and facilitation of cell growth and angiostatic effects. In addition, CXCL10 has been shown to be a significant biomarker of disease severity, and it can be used as a prognostic indicator for a variety of renal diseases, such as renal allograft dysfunction and lupus nephritis. In this review, we summarize the structures and biological functions of CXCL10 and CXCR3, focusing on the important role of CXCL10 in the pathogenesis of kidney disease, and provide a theoretical basis for CXCL10 as a potential biomarker and therapeutic target in human kidney disease.
Collapse
Affiliation(s)
- Jie Gao
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing 100853, China
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwu Road 324, Jinan 250000, China
| | - Lingling Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing 100853, China
| | - Siyang Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing 100853, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Beijing 100853, China
| |
Collapse
|
15
|
Abstract
Renal fibrosis is the final pathological process common to any ongoing, chronic kidney injury or maladaptive repair. Renal fibrosis is considered to be closely related to various cell types, such as fibroblasts, myofibroblasts, T cells, and other inflammatory cells. Multiple types of cells regulate renal fibrosis through the recruitment, proliferation, and activation of fibroblasts, and the production of the extracellular matrix. Cell trafficking is orchestrated by a family of small proteins called chemokines. Chemokines are cytokines with chemotactic properties, which are classified into 4 groups: CXCL, CCL, CX3CL, and XCL. Similarly, chemokine receptors are G protein-coupled seven-transmembrane receptors classified into 4 groups: XCR, CCR, CXCR, and CX3CR. Chemokine receptors are also implicated in the infiltration, differentiation, and survival of functional cells, triggering inflammation that leads to fibrosis development. In this review, we summarize the different chemokine receptors involved in the processes of fibrosis in different cell types. Further studies are required to identify the molecular mechanisms of chemokine signaling that contribute to renal fibrosis.
Collapse
|
16
|
Peddakkulappagari CS, Saifi MA, Khurana A, Anchi P, Singh M, Godugu C. Withaferin A ameliorates renal injury due to its potent effect on inflammatory signaling. Biofactors 2019; 45:750-762. [PMID: 31188510 DOI: 10.1002/biof.1534] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/22/2019] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is one of the major global health concerns and is responsible for end-stage renal disease (ESRD) complications. Inflammation plays a pivotal role in the progression of CKD. In the present study, we evaluated the renoprotective effects of a potent immunomodulator steroidal lactone, Withaferin A (WfA), in an animal model of renal injury (unilateral ureteral obstruction, UUO) and further investigated if the inhibition of inflammatory signaling can be a useful approach to reduce renal injury. Animals were randomly divided into five groups: Sham control, UUO control, WfA control, WfA low dose (1 mg/kg), and WfA high dose (3 mg/kg). Oxidative stress was measured by the estimation of reduced glutathione and lipid peroxidation levels. H&E and Picrosirius Red staining were performed to assess the extent of histological damage and collagen deposition. Furthermore, the molecular mechanism of the WfA effects was explored by immunohistochemistry, enzyme-linked immunosorbent assay, multiplex analysis of transforming growth factor β (TGF-β) pathway, and an array of inflammatory cytokines/chemokines. Interestingly, our pharmacological intervention significantly attenuated tissue collagen, inflammatory signaling, and macrophage signaling. WfA intervention abrogated the inflammatory signaling as evident from the modulated levels of chemokines and cytokines. The levels of TGF-β along with downstream signaling molecules were also attenuated by WfA treatment as revealed by inhibition in the expression of TGF-β1, TGF-β2, p-Smad2, p-Smad3, total Smad4, p-Akt, and p-ERK. We, to the best of our knowledge, prove for the first time that WfA has potential renoprotective activity against UUO-induced nephropathy due to its outstanding anti-inflammatory properties.
Collapse
Affiliation(s)
- Chandra S Peddakkulappagari
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Mohd A Saifi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Amit Khurana
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Pratibha Anchi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| |
Collapse
|
17
|
Abstract
Genetic variants in APOL1, encoding apolipoprotein L1, are major drivers of glomerular disease in peoples of sub-Saharan African descent. APOL1-associated primary glomerular diseases include focal segmental glomerulosclerosis, human immunodeficiency virus-associated nephropathies, and arterionephrosclerosis. Other conditions where APOL1 variants affect outcomes include membranous nephropathy, lupus nephritis, diabetic nephropathy, preeclampsia, and kidney transplant. In focal segmental glomerulosclerosis, APOL1 variants are associated with upregulation of RNA encoding chemokine C-X-C motif receptor 3 ligands and ubiquitin D; the significance of these findings remains unclear but may provide valuable insight into disease mechanisms.
Collapse
|
18
|
Reversing CXCL10 Deficiency Ameliorates Kidney Disease in Diabetic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2763-2773. [PMID: 30273603 DOI: 10.1016/j.ajpath.2018.08.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 08/04/2018] [Accepted: 08/23/2018] [Indexed: 12/11/2022]
Abstract
The excessive accumulation of extracellular matrix material in the kidney is a histopathologic hallmark of diabetic kidney disease that correlates closely with declining function. Although considerable research has focused on the role of profibrotic factors, comparatively little attention has been paid to the possibility that a diminution in endogenous antifibrotic factors may also contribute. Among the latter, the ELR- CXC chemokines, CXCL9, CXCL10, and CXCL11, have been shown to provide a stop signal to prevent excessive fibrosis. Although the plasma concentrations of CXCL9 and CXCL11 were similar, those of CXCL10 were markedly lower in diabetic db/db mice compared with control db/m mice. In cell culture, CXCL10 inhibited kidney fibroblast collagen production in response to high glucose and the prosclerotic growth factor, transforming growth factor-β. In vivo, recombinant murine CXCL10 reduced mesangial and peritubular matrix expansion, albuminuria, and glomerular hypertrophy in db/db mice. In bone marrow, a major source of circulating chemokines, the concentration of CXCL10 was lower in cells derived from diabetic mice than from their nondiabetic counterparts. Silencing of CXCR3, the cognate receptor for CXCL10, abrogated the antifibrotic effects of bone marrow-derived secretions. In conclusion, experimental diabetes is a state of CXCL10 deficiency and that restoration of CXCL10 abundance prevented fibrosis and the development of diabetic kidney disease in mice.
Collapse
|
19
|
Matsui M, Roche L, Geroult S, Soupé-Gilbert ME, Monchy D, Huerre M, Goarant C. Cytokine and Chemokine Expression in Kidneys during Chronic Leptospirosis in Reservoir and Susceptible Animal Models. PLoS One 2016; 11:e0156084. [PMID: 27219334 PMCID: PMC4878748 DOI: 10.1371/journal.pone.0156084] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 05/09/2016] [Indexed: 01/24/2023] Open
Abstract
Leptospirosis is caused by pathogenic spirochetes of the genus Leptospira. Humans can be infected after exposure to contaminated urine of reservoir animals, usually rodents, regarded as typical asymptomatic carriers of leptospires. In contrast, accidental hosts may present an acute form of leptospirosis with a range of clinical symptoms including the development of Acute Kidney Injury (AKI). Chronic Kidney Disease (CKD) is considered as a possible AKI-residual sequela but little is known about the renal pathophysiology consequent to leptospirosis infection. Herein, we studied the renal morphological alterations in relation with the regulation of inflammatory cytokines and chemokines, comparing two experimental models of chronic leptospirosis, the golden Syrian hamster that survived the infection, becoming carrier of virulent leptospires, and the OF1 mouse, a usual reservoir of the bacteria. Animals were monitored until 28 days after injection with a virulent L. borgpetersenii serogroup Ballum to assess chronic infection. Hamsters developed morphological alterations in the kidneys with tubulointerstitial nephritis and fibrosis. Grading of lesions revealed higher scores in hamsters compared to the slight alterations observed in the mouse kidneys, irrespective of the bacterial load. Interestingly, pro-fibrotic TGF-β was downregulated in mouse kidneys. Moreover, cytokines IL-1β and IL-10, and chemokines MIP-1α/CCL3 and IP-10/CXCL-10 were significantly upregulated in hamster kidneys compared to mice. These results suggest a possible maintenance of inflammatory processes in the hamster kidneys with the infiltration of inflammatory cells in response to bacterial carriage, resulting in alterations of renal tissues. In contrast, lower expression levels in mouse kidneys indicated a better regulation of the inflammatory response and possible resolution processes likely related to resistance mechanisms.
Collapse
Affiliation(s)
- Mariko Matsui
- Institut Pasteur International Network, Institut Pasteur de Nouvelle-Calédonie, Leptospirosis Research and Expertise Unit, Noumea, New Caledonia
| | - Louise Roche
- Institut Pasteur International Network, Institut Pasteur de Nouvelle-Calédonie, Leptospirosis Research and Expertise Unit, Noumea, New Caledonia
| | - Sophie Geroult
- Institut Pasteur International Network, Institut Pasteur de Nouvelle-Calédonie, Leptospirosis Research and Expertise Unit, Noumea, New Caledonia
| | - Marie-Estelle Soupé-Gilbert
- Institut Pasteur International Network, Institut Pasteur de Nouvelle-Calédonie, Leptospirosis Research and Expertise Unit, Noumea, New Caledonia
| | - Didier Monchy
- Anatomic Pathology Laboratory, Gaston-Bourret Territorial Hospital Center, Noumea, New Caledonia
| | - Michel Huerre
- Unité de Recherche et Expertise en Histotechnologie et Pathologie, Institut Pasteur, Paris, France.,Departement de Pathologie, Institut Curie, Paris, France
| | - Cyrille Goarant
- Institut Pasteur International Network, Institut Pasteur de Nouvelle-Calédonie, Leptospirosis Research and Expertise Unit, Noumea, New Caledonia
| |
Collapse
|
20
|
Liu G, Zhang W, Xiao Y, Lu P. Critical Role of IP-10 on Reducing Experimental Corneal Neovascularization. Curr Eye Res 2014; 40:891-901. [PMID: 25309995 DOI: 10.3109/02713683.2014.968934] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIM AND SCOPE To address the role of interferon-induced protein of 10 kDa (IP-10) in the course of corneal neovascularization (CrNV) in a mouse model of experimental corneal neovascularization. MATERIAL AND METHOD BALB/c mice that were 7- to 8-week-old male were included in the study. Corneal injury was induced by NaOH. Mice were randomly divided into 2 groups of IP-10 and vehicle. The alkali-treated eyes received 5 μl of 5 μg/ml IP-10 dissolved in 0.2% sodium hyaluronate for IP-10-treated group, or 5 μl of 0.2% sodium hyaluronate for vehicle-treated group twice a day for 7 days immediately after the alkali injury. 2 weeks after alkali injury, corneas were removed and used for whole mount CD31 staining. The percentages of neovascularization on corneal photographs were examined with digital image analysis. In other experiments, at indicated time intervals, the corneas were removed. Angiogenic factor expression in the early phase after injury was quantified by real-time PCR and western blot. The VEGF expression in macrophages infiltrating into burned corneas was examined by Flow cytometry (FCM) and immunofluorescence. Tube formation and cell proliferation of human retinal endothelial cells (HRECs) were detected after being stimulated with IP-10 in vitro. RESULTS The mRNA and protein expression of IP-10 and C-X-C motif chemokine receptor 3 (CXCR3) was augmented after the alkali injury (p < 0.05). Compared with vehicle-treated mice, IP-10-treated mice exhibited reduced CrNV 2 weeks after injury, as evidenced by diminished CD31-positive areas (p < 0.05). Concomitantly, the intracorneal mRNA and protein expression enhancement of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) was lower in IP-10-treated mice than in vehicle-treated mice after injury (p < 0.05). Moreover, IP-10 inhibited HREC tube formation and proliferation in vitro. CONCLUSION IP-10-treated mice exhibited reduced alkali-induced CrNV through decreasing intracorneal VEGF and bFGF expression, and inhibiting endothelial cell proliferation and tube formation.
Collapse
Affiliation(s)
- Gaoqin Liu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University , Suzhou , China and
| | | | | | | |
Collapse
|
21
|
Peng X, Xiao Z, Zhang J, Li Y, Dong Y, Du J. IL-17A produced by bothγδT and Th17 cells promotes renal fibrosis via RANTES-mediated leukocyte infiltration after renal obstruction. J Pathol 2014; 235:79-89. [PMID: 25158055 DOI: 10.1002/path.4430] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 08/06/2014] [Accepted: 08/17/2014] [Indexed: 01/11/2023]
Affiliation(s)
- Xiaogang Peng
- Beijing AnZhen Hospital; Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases. The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing China
| | - Zhicheng Xiao
- Beijing AnZhen Hospital; Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases. The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing China
| | - Jing Zhang
- Beijing AnZhen Hospital; Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases. The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing China
| | - Yulin Li
- Beijing AnZhen Hospital; Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases. The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing China
| | - Yanjun Dong
- Beijing AnZhen Hospital; Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases. The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing China
| | - Jie Du
- Beijing AnZhen Hospital; Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases. The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education; Beijing China
| |
Collapse
|
22
|
Xie H, Feng C, Fu Q, Sa YL, Xu YM. Crosstalk between TGF-β1 and CXCR3 signaling during urethral fibrosis. Mol Cell Biochem 2014; 394:283-90. [DOI: 10.1007/s11010-014-2104-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 05/15/2014] [Indexed: 10/25/2022]
|
23
|
Rybinski B, Franco-Barraza J, Cukierman E. The wound healing, chronic fibrosis, and cancer progression triad. Physiol Genomics 2014; 46:223-44. [PMID: 24520152 PMCID: PMC4035661 DOI: 10.1152/physiolgenomics.00158.2013] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/04/2014] [Indexed: 02/07/2023] Open
Abstract
For decades tumors have been recognized as "wounds that do not heal." Besides the commonalities that tumors and wounded tissues share, the process of wound healing also portrays similar characteristics with chronic fibrosis. In this review, we suggest a tight interrelationship, which is governed as a concurrence of cellular and microenvironmental reactivity among wound healing, chronic fibrosis, and cancer development/progression (i.e., the WHFC triad). It is clear that the same cell types, as well as soluble and matrix elements that drive wound healing (including regeneration) via distinct signaling pathways, also fuel chronic fibrosis and tumor progression. Hence, here we review the relationship between fibrosis and cancer through the lens of wound healing.
Collapse
Affiliation(s)
- Brad Rybinski
- Cancer Biology Program, Fox Chase Cancer Center/Temple Health, Philadelphia, Pennsylvania
| | | | | |
Collapse
|
24
|
Sahin H, Berres ML, Wasmuth HE. Therapeutic potential of chemokine receptor antagonists for liver disease. Expert Rev Clin Pharmacol 2014; 4:503-13. [DOI: 10.1586/ecp.11.24] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Chan CC, Cheng LY, Lu J, Huang YH, Chiou SH, Tsai PH, Huo TI, Lin HC, Lee FY. The role of interferon-γ inducible protein-10 in a mouse model of acute liver injury post induced pluripotent stem cells transplantation. PLoS One 2012; 7:e50577. [PMID: 23227188 PMCID: PMC3515611 DOI: 10.1371/journal.pone.0050577] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/23/2012] [Indexed: 12/11/2022] Open
Abstract
Background Liver injuries are important medical problems that require effective therapy. Stem cell or hepatocyte transplantation has the potential to restore function of the damaged liver and ameliorate injury. However, the regulatory factors crucial for the repair and regeneration after cell transplantation have not been fully characterized. Our study investigated the effects and the expression of the regulatory factors in mouse models of acute liver injury either transplanted with the induced pluripotent stem cells (iPS) or the hepatocytes that differentiated from iPS cells (iHL). Methods/Principal Findings Mice received CCl4 injection and were randomized to receive vehicle, iPS, or iHL transfusions vial tail veins and were observed for 24, 48 or 72 hours. The group of mice with iPS transplantation performed better than the group of mice receiving iHL in reducing the serum alanine aminotransferase, aspartate aminotransferase, and liver necrosis areas at 24 hours after CCl4 injury. Moreover, iPS significantly increased the numbers of proliferating hepatocytes at 48 hours. Cytokine array identified that chemokine IP-10 could be the potential regulatory factor that ameliorates liver injury. Further studies revealed that iPS secreted IP-10 in vitro and transfusion of iPS increased IP-10 protein and mRNA expressions in the injured livers in vivo. The primary hepatocytes and non-parenchyma cells were isolated from normal and injured livers. Hepatocytes from injured livers that received iPS treatment expressed more IP-10 mRNA than their non-hepatocyte counter-parts. In addition, animal studies revealed that administration of recombinant IP-10 (rIP-10) effectively reduced liver injuries while IP-10-neutralizing antibody attenuated the protective effects of iPS and decreased hepatocyte proliferation. Both iPS and rIP-10 significantly reduced the 72-hour mortality rate in mice that received multiple CCl4-injuries. Conclusions/Significance These findings suggested that IP-10 may have an important regulatory role in facilitating the repair and regeneration of injured liver after iPS transplantation.
Collapse
Affiliation(s)
- Che-Chang Chan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Division of Gastroenterology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Ling-Yi Cheng
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Division of Gastroenterology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Jean Lu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yi-Hsiang Huang
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Division of Gastroenterology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- * E-mail: (YH); (SC)
| | - Shih-Hwa Chiou
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Stem Cell Lab, Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- * E-mail: (YH); (SC)
| | - Ping-Hsing Tsai
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Stem Cell Lab, Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Teh-Ia Huo
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Division of Gastroenterology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Han-Chieh Lin
- Division of Gastroenterology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Fa-Yauh Lee
- Division of Gastroenterology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| |
Collapse
|
26
|
Sahin H, Wasmuth HE. Chemokines in tissue fibrosis. Biochim Biophys Acta Mol Basis Dis 2012; 1832:1041-8. [PMID: 23159607 DOI: 10.1016/j.bbadis.2012.11.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 11/06/2012] [Accepted: 11/08/2012] [Indexed: 12/20/2022]
Abstract
Fibrosis or scarring of diverse organs and tissues is considered as a pathologic consequence of a chronically altered wound healing response which is tightly linked to inflammation and angiogenesis. The recruitment of immune cells, local proliferation of fibroblasts and the consecutive accumulation of extracellular matrix proteins are common pathophysiological hallmarks of tissue fibrosis, irrespective of the organ involved. Chemokines, a family of chemotactic cytokines, appear to be central mediators of the initiation as well as progression of these biological processes. Traditionally chemokines have only been considered to play a critical role in orchestrating the influx of immune cells to sites of tissue injury. However, within the last years, further aspects of chemokine biology including fibroblast activation and angiogenesis have been deciphered in tissue fibrosis of many different organs. Interestingly, certain chemokines appear to mediate common effects in liver, kidney, lung, and skin of various animal models, while others mediate tissue specific effects. These aspects have to be kept in mind when extrapolating data of animal studies to early human trials. Nevertheless, the further understanding of chemokine effects in tissue fibrosis might be an attractive approach for identifying novel therapeutic targets in chronic organ damage associated with high morbidity and mortality. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Hacer Sahin
- Medical Department III, University Hospital Aachen, RWTH Aachen, Germany
| | | |
Collapse
|
27
|
Liang YJ, Luo J, Lu Q, Zhou Y, Wu HW, Zheng D, Ren YY, Sun KY, Wang Y, Zhang ZS. Gene profile of chemokines on hepatic stellate cells of schistosome-infected mice and antifibrotic roles of CXCL9/10 on liver non-parenchymal cells. PLoS One 2012; 7:e42490. [PMID: 22905138 PMCID: PMC3414521 DOI: 10.1371/journal.pone.0042490] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 07/09/2012] [Indexed: 12/12/2022] Open
Abstract
Hepatic stellate cells (HSCs) play a key role in the development of liver fibrosis caused by schistosomiasis. Chemokines were widely expressed and involved in cellular activation, proliferation and migration in inflammatory and infectious diseases. However, little is known about the expressions of chemokines on HSCs in the schistosoma infection. In addition, the roles of chemokines in pathogenesis of liver fibrosis are not totally clear. In our study, we used microarray to analyze the temporal gene expressions of primary HSCs isolated from mice with both acute and chronic schistosomiasis. Our microarray data showed that most of the chemokines expressed on HSCs were upregulated at 3 weeks post-infection (p.i) when the egg granulomatous response was not obviously evoked in the liver. However, some of them like CXCL9, CXCL10 and CXCL11 were subsequently decreased at 6 weeks p.i when the granulomatous response reached the peak. In the chronic stage, most of the differentially expressed chemokines maintained persistent high-abundances. Furthermore, several chemokines including CCR2, CCR5, CCR7, CXCR3, CXCR4, CCL2, CCL5, CCL21, CXCL9 and CXCL10 were expressed by HCSs and the abundances of them were changed following the praziquantel treatment in the chronic stage, indicating that chemokines were possibly necessary for the persistence of the chronic stage. In vitro experiments, hepatic non-parenchymal cells, primary HSCs and human HSCs line LX-2 were stimulated by chemokines. The results showed that CXCL9 and CXCL10, but not CXCL11 or CXCL4, significantly inhibited the gene expressions of Col1α1, Col3α1 and α-SMA, indicating the potential anti-fibrosis effect of CXCL9 and CXCL10 in schistosomiasis. More interestingly, soluble egg antigen (SEA) of Schistosoma japonicum was able to inhibit transcriptional expressions of some chemokines by LX-2 cells, suggesting that SEA was capable of regulating the expression pattern of chemokine family and modulating the hepatic immune microenvironment in schistosomiasis.
Collapse
Affiliation(s)
- Yue-jin Liang
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Luo
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiao Lu
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Zhou
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hai-wei Wu
- Center for International Health Research, Rhode Island Hospital, Brown University, Providence, Rhode Island, United States of America
| | - Dan Zheng
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong-ya Ren
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ke-yi Sun
- Department of Microbiology & Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong Wang
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- * E-mail:
| | - Zhao-song Zhang
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
28
|
Sahin H, Borkham-Kamphorst E, Kuppe C, Zaldivar MM, Grouls C, Al-samman M, Nellen A, Schmitz P, Heinrichs D, Berres ML, Doleschel D, Scholten D, Weiskirchen R, Moeller MJ, Kiessling F, Trautwein C, Wasmuth HE. Chemokine Cxcl9 attenuates liver fibrosis-associated angiogenesis in mice. Hepatology 2012; 55:1610-9. [PMID: 22237831 DOI: 10.1002/hep.25545] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Recent data suggest that the chemokine receptor CXCR3 is functionally involved in fibroproliferative disorders, including liver fibrosis. Neoangiogenesis is an important pathophysiological feature of liver scarring, but a functional role of angiostatic CXCR3 chemokines in this process is unclear. We therefore investigated neoangiogenesis in carbon tetrachloride (CCl(4))-induced liver fibrosis in Cxcr3(-/-) and wildtype mice by histological, molecular, and functional imaging methods. Furthermore, we assessed the direct role of vascular endothelial growth factor (VEGF) overexpression on liver angiogenesis and the fibroproliferative response using a Tet-inducible bitransgenic mouse model. The feasibility of attenuation of angiogenesis and associated liver fibrosis by therapeutic treatment with the angiostatic chemokine Cxcl9 was systematically analyzed in vitro and in vivo. The results demonstrate that fibrosis progression in Cxcr3(-/-) mice was strongly linked to enhanced neoangiogenesis and VEGF/VEGFR2 expression compared with wildtype littermates. Systemic VEGF overexpression led to a fibrogenic response within the liver and was associated with a significantly increased Cxcl9 expression. In vitro, Cxcl9 displayed strong antiproliferative and antimigratory effects on VEGF-stimulated endothelial cells and stellate cells by way of reduced VEGFR2 (KDR), phospholipase Cγ (PLCγ), and extracellular signal-regulated kinase (ERK) phosphorylation, identifying this chemokine as a direct counter-regulatory molecule of VEGF signaling within the liver. Accordingly, systemic administration of Cxcl9 led to a strong attenuation of neoangiogenesis and experimental liver fibrosis in vivo. CONCLUSION The results identify direct angiostatic and antifibrotic effects of the Cxcr3 ligand Cxcl9 in a model of experimental liver fibrosis. The amelioration of liver damage by systemic application of Cxcl9 might offer a novel therapeutic approach for chronic liver diseases associated with increased neoangiogenesis.
Collapse
Affiliation(s)
- Hacer Sahin
- Medical Department III, University Hospital Aachen, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Casrouge A, Bisiaux A, Stephen L, Schmolz M, Mapes J, Pfister C, Pol S, Mallet V, Albert ML. Discrimination of agonist and antagonist forms of CXCL10 in biological samples. Clin Exp Immunol 2012; 167:137-48. [PMID: 22132893 DOI: 10.1111/j.1365-2249.2011.04488.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The ready access to commercially available multiplex assays and the importance of inflammation in disease pathogenesis has resulted in an abundance of studies aimed at identifying surrogate biomarkers for different clinically important questions. Establishing a link between a biomarker and disease pathogenesis, however, is quite complex, and in some instances this complexity is compounded by post-translational modifications and the use of immunoassays that do not always discriminate between the different forms of the same protein. Herein, we provide a detailed description of an assay system that has been established to discriminate the agonist form of CXCL10 from the NH(2) -terminal truncated form of the molecule generated by dipeptidylpeptidase IV (DPP4) cleavage. We demonstrate the utility of this assay system for monitoring agonist and antagonist forms of CXCL10 in culture supernatant, patient plasma and urine samples. Given the important role of CXCL10 in chronic inflammatory diseases and its suggested role as a predictive marker in managing patients with chronic hepatitis C, asthma, atopic dermatitis, transplantation, tuberculosis, kidney injury, cancer and other diseases, we believe that our method will be of general interest to the research and medical community.
Collapse
Affiliation(s)
- A Casrouge
- Laboratory of Dendritic Cell Biology, Department of Immunology, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Chemokines are a class of small cytokine-like molecules that orchestrate immune cell infiltration into the liver in response to acute and chronic injuries. Apart from their chemotactic effect, however, chemokines seem to mediate many other aspects of liver diseases, including a direct activation of stellate cells, the modulation of hepatocyte proliferation and angiogenesis. The identification of specific biological functions for chemokines in liver diseases has been hampered by the finding that resident and infiltrating cells in the liver are often a source, as well as a target, of chemokines. Furthermore, chemokines might cause differing effects depending on the etiology of liver damage, their local concentrations and their ability to form multimers and heterodimers. Nevertheless, the functions of a number of important chemokines and their associated receptors have been identified in both in vivo and in vitro studies. Indeed, harmful (proinflammatory, profibrogenic) and beneficial (antifibrogenic, antiangiogenic) effects of chemokines have been discovered in experimental liver disease models. In this Review, the current knowledge of chemokines in experimental liver disease models is summarized. Advances that might lead to preclinical applications are discussed, as are the roles of chemokine receptors as promising pharmacologically targetable molecules.
Collapse
|
31
|
Grande MT, Pérez-Barriocanal F, López-Novoa JM. Role of inflammation in túbulo-interstitial damage associated to obstructive nephropathy. JOURNAL OF INFLAMMATION-LONDON 2010; 7:19. [PMID: 20412564 PMCID: PMC2873503 DOI: 10.1186/1476-9255-7-19] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 04/22/2010] [Indexed: 02/07/2023]
Abstract
Obstructive nephropathy is characterized by an inflammatory state in the kidney, that is promoted by cytokines and growth factors produced by damaged tubular cells, infiltrated macrophages and accumulated myofibroblasts. This inflammatory state contributes to tubular atrophy and interstitial fibrosis characteristic of obstructive nephropathy. Accumulation of leukocytes, especially macrophages and T lymphocytes, in the renal interstitium is strongly associated to the progression of renal injury. Proinflammatory cytokines, NF-κB activation, adhesion molecules, chemokines, growth factors, NO and oxidative stress contribute in different ways to progressive renal damage induced by obstructive nephropathy, as they induce leukocytes recruitment, tubular cell apoptosis and interstitial fibrosis. Increased angiotensin II production, increased oxidative stress and high levels of proinflammatory cytokines contribute to NF-κB activation which in turn induce the expression of adhesion molecules and chemokines responsible for leukocyte recruitment and iNOS and cytokines overexpression, which aggravates the inflammatory response in the damaged kidney. In this manuscript we revise the different events and regulatory mechanisms involved in inflammation associated to obstructive nephropathy.
Collapse
Affiliation(s)
- María T Grande
- Instituto "Reina Sofía" de Investigación Nefrológica, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain.
| | | | | |
Collapse
|
32
|
Affiliation(s)
- Jeffrey M Davidson
- Department of Pathology, Vanderbilt University School of Medicine, Nashville, TN 37232-2562, USA.
| |
Collapse
|
33
|
Antifibrotic effects of CXCL9 and its receptor CXCR3 in livers of mice and humans. Gastroenterology 2009; 137:309-19, 319.e1-3. [PMID: 19344719 PMCID: PMC2892869 DOI: 10.1053/j.gastro.2009.03.053] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 03/16/2009] [Accepted: 03/25/2009] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Fibrosis is the hallmark of chronic liver diseases, yet many aspects of its mechanism remain to be defined. Chemokines are ubiquitous chemotactic molecules that mediate many acute and chronic inflammatory conditions, and CXC chemokine genes colocalize with a locus previously shown to include fibrogenic genes. We investigated the roles of the chemokine CXCL9 and its receptor CXCR3 in liver fibrosis. METHODS The effects of CXCL variants on fibrogenesis were analyzed using samples from patients with hepatitis C virus infection and by induction of fibrosis in CXCR3(-/-) and wild-type mice. In mice, intrahepatic immune cell subsets were investigated and interferon gamma messenger RNA levels were measured at baseline and after injury. Human serum CXCL9 levels were measured and correlated with CXCL9 variant and fibrosis severity. The effects of stimulation with CXCL9 were investigated on human hepatic stellate cells (LX-2). RESULTS Specific CXCL9 variants were associated with liver fibrosis in mice and humans; CXCL9 serum concentrations correlated with genotypes and levels of fibrosis in patients. In contrast to other chemokines, CXCL9 exerted antifibrotic effects in vitro, suppressing collagen production in LX-2 cells. CXCR3(-/-) mice had increased liver fibrosis; progression was associated with decreased numbers of intrahepatic interferon gamma-positive T cells and reduced interferon gamma messenger RNA, indicating that CXCL9-CXCR3 regulates Th1-associated immune pathways. CONCLUSIONS This is the first description of a chemokine-based antifibrotic pathway in the liver; antifibrotic therapies might be developed to modulate CXC chemokine levels.
Collapse
|
34
|
Kapoor S. Emerging new chemokine CXCR3 antagonists and their potential clinical and therapeutic applications. Hepatology 2009; 49:1400-1. [PMID: 19330874 DOI: 10.1002/hep.22781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
35
|
Okamura DM, Pennathur S, Pasichnyk K, López-Guisa JM, Collins S, Febbraio M, Heinecke J, Eddy AA. CD36 regulates oxidative stress and inflammation in hypercholesterolemic CKD. J Am Soc Nephrol 2009; 20:495-505. [PMID: 19211715 DOI: 10.1681/asn.2008010009] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Scavenger receptors play a central role in atherosclerosis by processing oxidized lipoproteins and mediating their cellular effects. Recent studies suggested that the atherogenic state correlates with progression of chronic kidney disease (CKD); therefore, scavenger receptors are candidate mediators of renal fibrogenesis. Here, we investigated the role of CD36, a class B scavenger receptor, in a hypercholesterolemic model of CKD. We placed CD36-deficient mice and wild-type male mice on a high-fat Western diet for 7 to 8 wk and then performed either sham or unilateral ureteral obstruction surgery. CD36-deficient mice developed significantly less fibrosis compared with wild-type mice at days 3, 7, and 14 after obstruction. Compared with wild-type mice, CD36-deficient mice had significantly more interstitial macrophages at 7 d but not at 14 d. CD36-deficient mice exhibited reduced levels of activated NF-kappaB and oxidative stress (assessed by measuring fatty acid-derived hydroxyoctadecadienoic acid and protein carbonyl content) and decreased accumulation of interstitial myofibroblasts compared with wild-type mice. These data suggest that CD36 is a key modulator of proinflammatory and oxidative pathways that promote fibrogenesis in CKD.
Collapse
Affiliation(s)
- Daryl M Okamura
- Department of Pediatrics, Seattle Children's Research Institute, Division of Nephrology, Seattle, WA 98105, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hsu HH, Duning K, Meyer HH, Stölting M, Weide T, Kreusser S, van Le T, Gerard C, Telgmann R, Brand-Herrmann SM, Pavenstädt H, Bek MJ. Hypertension in mice lacking the CXCR3 chemokine receptor. Am J Physiol Renal Physiol 2009; 296:F780-9. [PMID: 19129260 DOI: 10.1152/ajprenal.90444.2008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The CXC chemokine receptor 3 (CXCR3) has been linked to autoimmune and inflammatory disease, allograft rejection, and ischemic nephropathy. CXCR3 is expressed on endothelial and smooth muscle cells. Although a recent study posited that antagonizing of CXCR3 function may reduce atherosclerosis, the role of CXCR3 in controlling physiological vascular functions remains unclear. This study demonstrates that disruption of CXCR3 leads to elevated mean arterial pressures in anesthetized and conscious mice, respectively. Stimulation of isolated resistance vessels with various vasoconstrictors showed increased contractibility in CXCR3-/- mice in response to angiotensin II (ANG II) and a decreased vasodilatation in response to acetylcholine (ACh). The increased contractibility was related to higher ANG II type 1 receptor (AT1R) expression, whereas the decreased vasodilatation was related to lower M3-ACh receptor expression in the mesenteric arteries of CXCR3-/- mice compared with wild-type mice. The vasodilatatory response to ACh could be antagonized by the nonselective ACh receptor antagonist atropine and the selective M3 receptor antagonist 4-DAMP, but not by M1, M2, and M4 receptor antagonists. Additionally, EMSA studies revealed that transcription factors SP-1 and EGR-1 interact as a complex with the murine AT1R promoter region. Furthermore, we could show increased expression of SP-1 in CXCR3-/- mice indicating an imbalanced SP-1 and EGR-1 complex formation which causes increased AT1R expression and hypertension. The data indicate that CXCR3 receptor is important in vascular contractility and hypertension, possibly through upregulated AT1R expression.
Collapse
Affiliation(s)
- Hsiang-Hao Hsu
- Dept. of Internal Medicine, Albert-Schweitzer-Str. 33, D-48149 Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|