1
|
Cao Q, Foley M, Gill AJ, Chou A, Chen XM, Pollock CA. Second Generation I-Body AD-214 Attenuates Unilateral Ureteral Obstruction (UUO)-Induced Kidney Fibrosis Through Inhibiting Leukocyte Infiltration and Macrophage Migration. Int J Mol Sci 2024; 25:13127. [PMID: 39684834 DOI: 10.3390/ijms252313127] [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: 11/06/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Kidney fibrosis is the common pathological pathway in progressive chronic kidney disease (CKD), and current treatments are largely ineffective. The C-X-C chemokine receptor 4 (CXCR4) is crucial to fibrosis development. By using neural cell adhesion molecules as scaffolds with binding loops that mimic the shape of shark antibodies, fully humanized single-domain i-bodies have been developed. The first-generation i-body, AD-114, demonstrated antifibrotic effects in a mouse model of folic acid (FA)-induced renal fibrosis. The second-generation i-body, AD-214, is an Fc-fusion protein with an extended half-life, enhanced activity, and a mutated Fc domain to prevent immune activation. To investigate the renoprotective mechanisms of AD-214, RPTEC/TERT1 cells (a human proximal tubular cell line) were incubated with TGF-b1 with/without AD-214 and the supernatant was collected to measure collagen levels by Western blot. Mice with unilateral ureteral obstruction (UUO) received AD-214 intraperitoneally (i.p.) every two days for 14 days. Kidney fibrosis markers and kidney function were then analyzed. AD-214 suppressed TGF-b1-induced collagen overexpression in RPTEC/TERT1 cells. In UUO mice, AD-214 reduced extracellular matrix (ECM) deposition, restored kidney function, and limited leukocyte infiltration. In a scratch assay, AD-214 also inhibited macrophage migration. To conclude, i-body AD-214 attenuates UUO-induced kidney fibrosis by inhibiting leukocyte infiltration and macrophage migration.
Collapse
Affiliation(s)
- Qinghua Cao
- Renal Medicine, Kolling Institute of Medical Research, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Michael Foley
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
- AdAlta Limited, LIMS2 Building, Science Drive, La Trobe University, Melbourne, VIC 3086, Australia
| | - Anthony J Gill
- Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, Sydney, NSW 2065, Australia
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
| | - Angela Chou
- Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, Sydney, NSW 2065, Australia
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW 2064, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
| | - Xin-Ming Chen
- Renal Medicine, Kolling Institute of Medical Research, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Carol A Pollock
- Renal Medicine, Kolling Institute of Medical Research, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| |
Collapse
|
2
|
Huang Y, Li L, Chen R, Yu L, Zhao S, Jia Y, Dou Y, Zhang Z, An Y, Tang X, Zhao X, Zhou L. Heterogeneous phenotype of a Chinese Familial WHIM syndrome with CXCR4 V340fs gain-of-function mutation. Front Immunol 2024; 15:1460990. [PMID: 39575248 PMCID: PMC11578956 DOI: 10.3389/fimmu.2024.1460990] [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: 07/07/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Background WHIM syndrome is a rare, autosomal dominant inborn error of immunity characterized by warts, hypogammaglobulinemia, infection, and myelokathexis. It is caused mainly by heterozygous mutations at the C-terminus of the C-X-C chemokine receptor type 4 (CXCR4) gene. Methods We described the detailed clinical, genetic, immunological and treatment characteristic of four WHIM patients from a single Chinese family. Results Here, we report four patients from a family carrying a variant of CXCR4 (c.1016_1017dupCT), which introduces a frameshift at codon V340, resulting in an extension of 14 amino acids (p.V340L fs*27). We provide and in-depth analysis of their clinical, genetic, immunological and treatment characteristic, noting that these patients exhibited an atypical clinical phenotype when compared to reported CXCR4R334X patients. Additionally, the frameshift variant CXCR4V340fs led to impaired receptor downregulation in patients' PBMCs, and in HEK293T cells transfected with the variant plasmids. Conclusions Our study provided detailed clinical features of four CXCR4V340fs WHIM patients from one Chinese family who presented atypical phenotype and enrich the spectrum of WHIM syndrome.
Collapse
Affiliation(s)
- Yu Huang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Hematology Oncology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lu Li
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Ran Chen
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lang Yu
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Shunkai Zhao
- Department of Biology, School of Arts and Sciences, Tufts University, Medford, MA, United States
| | - Yanjun Jia
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Dou
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Hematology Oncology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyong Zhang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Rheumatism and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Yunfei An
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Rheumatism and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Tang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Rheumatism and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Zhao
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Rheumatism and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lina Zhou
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Li C, Chen J, Chen Y, Zhang C, Yang H, Yu S, Song H, Fu P, Zeng X. The association between patterns of exposure to adverse life events and the risk of chronic kidney disease: a prospective cohort study of 140,997 individuals. Transl Psychiatry 2024; 14:424. [PMID: 39375339 PMCID: PMC11458756 DOI: 10.1038/s41398-024-03114-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
Exposure to adverse life events is linked to somatic disorders. The study aims to evaluate the association between adverse events at varying life stages and the risk of chronic kidney disease (CKD), a condition affecting about 10% population worldwide. This prospective cohort study included 140,997 participants from the UK Biobank. Using survey items related to childhood maltreatment, adulthood adversity and catastrophic trauma, we performed latent class analysis to summarize five distinct patterns of exposure to adverse life events, namely "low-level exposure", "childhood exposure", "adulthood exposure", "sexual abuse" and "child-to-adulthood exposure". We used Cox proportional hazard regression to evaluate the association of patterns of exposure to adverse life events with CKD, regression-based mediation analysis to decompose the total effect, and gene-environment-wide interaction study (GEWIS) to identify interactions between genetic loci and adverse life events. During a median follow-up of 5.98 years, 2734 cases of incident CKD were identified. Compared with the "low-level exposure" pattern, "child-to-adulthood exposure" was associated with increased risk of CKD (hazard ratio 1.37, 95% CI 1.14 to 1.65). BMI, smoking and hypertension mediated 11.45%, 9.79%, and 4.50% of this total effect, respectively. Other patterns did not show significant results. GEWIS and subsequent analyses indicated that the magnitude of the association between adverse life events and CKD differed according to genetic polymorphisms, and identified potential underlying pathways (e.g., interleukin 1 receptor activity). These findings underscore the importance of incorporating an individual's psychological encounters and genetic profiles into the precision prevention of CKD.
Collapse
Affiliation(s)
- Chunyang Li
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Chen
- Central Laboratory, Sichuan Academy of Medical Science and Sichuan Provincial Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yilong Chen
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Zhang
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huazhen Yang
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shaobin Yu
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huan Song
- Center of Mental Health, West China Hospital, Sichuan University, Chengdu, China
- Center of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Ping Fu
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Scharf P, Sandri S, Rizzetto F, Xavier LF, Grosso D, Correia-Silva RD, Farsky PS, Gil CD, Farsky SHP. GPCRs overexpression and impaired fMLP-induced functions in neutrophils from chronic kidney disease patients. Front Immunol 2024; 15:1387566. [PMID: 39253088 PMCID: PMC11381270 DOI: 10.3389/fimmu.2024.1387566] [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: 02/18/2024] [Accepted: 08/02/2024] [Indexed: 09/11/2024] Open
Abstract
Introduction G-protein coupled receptors (GPCRs) expressed on neutrophils regulate their mobilization from the bone marrow into the blood, their half-live in the circulation, and their pro- and anti-inflammatory activities during inflammation. Chronic kidney disease (CKD) is associated with systemic inflammatory responses, and neutrophilia is a hallmark of CKD onset and progression. Nonetheless, the role of neutrophils in CKD is currently unclear. Methods Blood and renal tissue were collected from non-dialysis CKD (grade 3 - 5) patients to evaluate GPCR neutrophil expressions and functions in CKD development. Results CKD patients presented a higher blood neutrophil-to-lymphocyte ratio (NLR), which was inversely correlated with the glomerular filtration rate (eGFR). A higher frequency of neutrophils expressing the senescent GPCR receptor (CXCR4) and activation markers (CD18+CD11b+CD62L+) was detected in CKD patients. Moreover, CKD neutrophils expressed higher amounts of GPCR formyl peptide receptors (FPR) 1 and 2, known as neutrophil pro- and anti-inflammatory receptors, respectively. Cytoskeletal organization, migration, and production of reactive oxygen species (ROS) by CKD neutrophils were impaired in response to the FPR1 agonist (fMLP), despite the higher expression of FPR1. In addition, CKD neutrophils presented enhanced intracellular, but reduced membrane expression of the protein Annexin A1 (AnxA1), and an impaired ability to secrete it into the extracellular compartment. Secreted and phosphorylated AnxA1 is a recognized ligand of FPR2, pivotal in anti-inflammatory and efferocytosis effects. CKD renal tissue presented a low number of neutrophils, which were AnxA1+. Conclusion Together, these data highlight that CKD neutrophils overexpress GPCRs, which may contribute to an unbalanced aging process in the circulation, migration into inflamed tissues, and efferocytosis.
Collapse
Affiliation(s)
- Pablo Scharf
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Felipe Rizzetto
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Lagoa Federal Hospital, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luana Filippi Xavier
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Rebeca D Correia-Silva
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Pedro S Farsky
- Dante Pazzanese Institute of Cardiology of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Cristiane D Gil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
Garcia NA, Gonzalez-King H, Mellergaard M, Nair S, Salomon C, Handberg A. Comprehensive strategy for identifying extracellular vesicle surface proteins as biomarkers for chronic kidney disease. Front Physiol 2024; 15:1328362. [PMID: 38379702 PMCID: PMC10877036 DOI: 10.3389/fphys.2024.1328362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Chronic kidney disease (CKD) poses a significant health burden worldwide. Especially, obesity-induced chronic kidney disease (OCKD) is associated with a lack of accuracy in disease diagnostic methods. The identification of reliable biomarkers for the early diagnosis and monitoring of CKD and OCKD is crucial for improving patient outcomes. Extracellular vesicles (EVs) have emerged as potential biomarkers in the context of CKD. In this review, we focused on the role of EVs as potential biomarkers in CKD and OCKD and developed a comprehensive list of EV membrane proteins that could aid in the diagnosis and monitoring of the disease. To assemble our list, we employed a multi-step strategy. Initially, we conducted a thorough review of the literature on EV protein biomarkers in kidney diseases. Additionally, we explored papers investigating circulating proteins as biomarkers in kidney diseases. To further refine our list, we utilized the EV database Vesiclepedia.org to evaluate the qualifications of each identified protein. Furthermore, we consulted the Human Protein Atlas to assess the localization of these candidates, with a particular focus on membrane proteins. By integrating the information from the reviewed literature, Vesiclepedia.org, and the Human Protein Atlas, we compiled a comprehensive list of potential EV membrane protein biomarkers for CKD and OCKD. Overall, our review underscores the potential of EVs as biomarkers in the field of CKD research, providing a foundation for future studies aimed at improving CKD and OCKD diagnosis and treatment.
Collapse
Affiliation(s)
| | - Hernan Gonzalez-King
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maiken Mellergaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, The Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland, Brisbane, QLD, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland, Brisbane, QLD, Australia
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, The Faculty of Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
6
|
Wu Q, Chen Q, Xu D, Wang X, Ye H, Li X, Xiong Y, Li J, Zhou S, Miao J, Shen W, Liu Y, Niu H, Tang Y, Zhou L. C-X-C chemokine receptor type 4 promotes tubular cell senescence and renal fibrosis through β-catenin-inhibited fatty acid oxidation. J Cell Mol Med 2024; 28:e18075. [PMID: 38213100 PMCID: PMC10844696 DOI: 10.1111/jcmm.18075] [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: 03/31/2023] [Revised: 11/01/2023] [Accepted: 11/24/2023] [Indexed: 01/13/2024] Open
Abstract
The prevalence of chronic kidney disease (CKD) is highly increasing. Renal fibrosis is a common pathological feature in various CKD. Previous studies showed tubular cell senescence is highly involved in the pathogenesis of renal fibrosis. However, the inducers of tubular senescence and the underlying mechanisms have not been fully investigated. C-X-C motif chemokine receptor 4 (CXCR4), a G-protein-coupled seven-span transmembrane receptor, increases renal fibrosis and plays an important role in tubular cell injury. Whereas, whether CXCR4 could induce tubular cell senescence and the detailed mechanisms have not studied yet. In this study, we adopted adriamycin nephropathy and 5/6 nephrectomy models, and cultured tubular cell line. Overexpression or knockdown of CXCR4 was obtained by injection of related plasmids. We identified CXCR4 increased in injury tubular cells. CXCR4 was expressed predominantly in renal tubular epithelial cells and co-localized with adipose differentiation-related protein (ADRP) as well as the senescence-related protein P16INK4A . Furthermore, we found overexpression of CXCR4 greatly induced the activation of β-catenin, while knockdown of CXCR4 inhibited it. We also found that CXCR4 inhibited fatty acid oxidation and triggered lipid deposition in tubular cells. To inhibit β-catenin by ICG-001, an inhibitor of β-catenin, could significantly block CXCR4-suppressed fatty acid oxidation. Taken together, our results indicate that CXCR4 is a key mediator in tubular cell senescence and renal fibrosis. CXCR4 promotes tubular cell senescence and renal fibrosis by inducing β-catenin and inhibiting fatty acid metabolism. Our findings provide a new theory for tubular cell injury in renal fibrosis.
Collapse
Affiliation(s)
- Qinyu Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of NephrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Qiurong Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Dan Xu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaoxu Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Huiyun Ye
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaolong Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yabing Xiong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jiemei Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Weiwei Shen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hongxin Niu
- Special Medical Service Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ying Tang
- Department of NephrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
Wu Q, Zhou S, Xu D, Meng P, Chen Q, Wang X, Li X, Chen S, Ye H, Ye W, Xiong Y, Li J, Miao J, Shen W, Lin X, Hou FF, Liu Y, Zhang Y, Zhou L. The CXCR4-AT1 axis plays a vital role in glomerular injury via mediating the crosstalk between podocyte and mesangial cell. Transl Res 2024; 264:15-32. [PMID: 37696390 DOI: 10.1016/j.trsl.2023.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 08/18/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023]
Abstract
Glomeruli stand at the center of nephrons to accomplish filtration and albumin interception. Podocytes and mesangial cells are the major constituents in the glomeruli. However, their interdependency in glomerular injury has rarely been reported. Herein, we investigated the role of C-X-C chemokine receptor type 4 (CXCR4) in mediating the crosstalk between podocytes and mesangial cells. We found CXCR4 and angiotensin II (AngII) increased primarily in injured podocytes. However, type-1 receptor of angiotensin II (AT1) and stromal cell-derived factor 1α (SDF-1α), a ligand of CXCR4, were evidently upregulated in mesangial cells following the progression of podocyte injury. Ectopic expression of CXCR4 in 5/6 nephrectomy mice increased the decline of renal function and glomerular injury, accelerated podocyte injury and mesangial cell activation, and initiated CXCR4-AT1 axis signals. Additionally, treatment with losartan, an AT1 blocker, interrupted the cycle of podocyte injury and mesangial matrix deposition triggered by CXCR4. Podocyte-specific ablation of CXCR4 gene blocked podocyte injury and mesangial cell activation. In vitro, CXCR4 overexpression induced oxidative stress and renin angiotensin system (RAS) activation in podocytes, and triggered the communication between podocytes and mesangial cells. In cultured mesangial cells, AngII treatment induced the expression of SDF-1α, which was secreted into the supernatant to further promote oxidative stress and cell injury in podocytes. Collectively, these results demonstrate that the CXCR4-AT1 axis plays a vital role in glomerular injury via mediating pathologic crosstalk between podocytes and mesangial cells. Our findings uncover a novel pathogenic mechanism by which the CXCR4-AT1 axis promotes glomerular injury.
Collapse
Affiliation(s)
- Qinyu Wu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Shan Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Dan Xu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Ping Meng
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Qiurong Chen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Xiaoxu Wang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Xiaolong Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Shuangqin Chen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Huiyun Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Wenting Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Yabing Xiong
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Jiemei Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Jinhua Miao
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Weiwei Shen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Xu Lin
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Yunfang Zhang
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.
| |
Collapse
|
8
|
Szóstek-Mioduchowska A, Wójtowicz A, Sadowska A, Moza Jalali B, Słyszewska M, Łukasik K, Gurgul A, Szmatoła T, Bugno-Poniewierska M, Ferreira-Dias G, Skarzynski DJ. Transcriptomic profiling of mare endometrium at different stages of endometrosis. Sci Rep 2023; 13:16263. [PMID: 37758834 PMCID: PMC10533846 DOI: 10.1038/s41598-023-43359-5] [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: 08/07/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023] Open
Abstract
In the current study, transcriptome profiles of mare endometrium, classified into categories I, IIA, and IIB according to Kenney and Doig, were compared using RNA sequencing, analyzed, and functionally annotated using in silico analysis. In the mild stage (IIA) of endometrosis compared to category I endometrium, differentially expressed genes (DEGs) were annotated to inflammation, abnormal metabolism, wound healing, and quantity of connective tissue. In the moderate stage (IIB) of endometrosis compared to category I endometrium, DEGs were annotated to inflammation, fibrosis, cellular homeostasis, mitochondrial dysfunction, and pregnancy disorders. Ingenuity pathway analysis (IPA) identified cytokines such as transforming growth factor (TGF)-β1, interleukin (IL)-4, IL-13, and IL-17 as upstream regulators of DEGs associated with cellular homeostasis, metabolism, and fibrosis signaling pathways. In vitro studies showed the effect of these cytokines on DEGs such as ADAMTS1, -4, -5, -9, and HK2 in endometrial fibroblasts at different stages of endometrosis. The effect of cytokines on ADAMTS members' gene transcription in fibroblasts differs according to the severity of endometrosis. The identified transcriptomic changes associated with endometrosis suggest that inflammation and metabolic changes are features of mild and moderate stages of endometrosis. The changes of ADAMTS-1, -4, -5, -9, in fibrotic endometrium as well as in endometrial fibroblast in response to TGF-β1, IL-4, IL-13, and IL-17 suggest the important role of these factors in the development of endometrosis.
Collapse
Affiliation(s)
- A Szóstek-Mioduchowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research Polish Academy of Sciences in Olsztyn, Olsztyn, Poland.
| | - A Wójtowicz
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - A Sadowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - B Moza Jalali
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - M Słyszewska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - K Łukasik
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| | - A Gurgul
- Center for Experimental and Innovative Medicine, University of Agriculture in Cracow, Cracow, Poland
| | - T Szmatoła
- Center for Experimental and Innovative Medicine, University of Agriculture in Cracow, Cracow, Poland
| | - M Bugno-Poniewierska
- Department of Animal Reproduction, Anatomy and Genomics, University of Agriculture in Cracow, Cracow, Poland
| | - G Ferreira-Dias
- Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - D J Skarzynski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research Polish Academy of Sciences in Olsztyn, Olsztyn, Poland
| |
Collapse
|
9
|
Khalil NN, Petersen AP, Song CJ, Chen Y, Takamoto K, Kellogg AC, Chen EZ, McMahon AP, McCain ML. User-friendly microfluidic system reveals native-like morphological and transcriptomic phenotypes induced by shear stress in proximal tubule epithelium. APL Bioeng 2023; 7:036106. [PMID: 37584027 PMCID: PMC10424157 DOI: 10.1063/5.0143614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
Drug-induced nephrotoxicity is a leading cause of drug attrition, partly due to the limited relevance of pre-clinical models of the proximal tubule. Culturing proximal tubule epithelial cells (PTECs) under fluid flow to mimic physiological shear stress has been shown to improve select phenotypes, but existing flow systems are expensive and difficult to implement by non-experts in microfluidics. Here, we designed and fabricated an accessible and modular flow system for culturing PTECs under physiological shear stress, which induced native-like cuboidal morphology, downregulated pathways associated with hypoxia, stress, and injury, and upregulated xenobiotic metabolism pathways. We also compared the expression profiles of shear-dependent genes in our in vitro PTEC tissues to that of ex vivo proximal tubules and observed stronger clustering between ex vivo proximal tubules and PTECs under physiological shear stress relative to PTECs under negligible shear stress. Together, these data illustrate the utility of our user-friendly flow system and highlight the role of shear stress in promoting native-like morphological and transcriptomic phenotypes in PTECs in vitro, which is critical for developing more relevant pre-clinical models of the proximal tubule for drug screening or disease modeling.
Collapse
Affiliation(s)
- Natalie N. Khalil
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Andrew P. Petersen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | | | - Yibu Chen
- USC Libraries Bioinformatics Service, University of Southern California, Los Angeles, California 90089, USA
| | - Kaelyn Takamoto
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Austin C. Kellogg
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Elaine Zhelan Chen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | | | - Megan L. McCain
- Author to whom correspondence should be addressed:. Tel.: +1 2138210791. URL:https://livingsystemsengineering.usc.edu
| |
Collapse
|
10
|
Dutta A, Saha S, Bahl A, Mittal A, Basak T. A comprehensive review of acute cardio-renal syndrome: need for novel biomarkers. Front Pharmacol 2023; 14:1152055. [PMID: 37288107 PMCID: PMC10242013 DOI: 10.3389/fphar.2023.1152055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/03/2023] [Indexed: 06/09/2023] Open
Abstract
Cardiorenal syndrome represents a wide-spectrum disorder involving the heart and kidneys as the primary affected organs. India has an increasingly high burden of acute CRS, coinciding with the rise in global statistics. Up to 2022, approximately 46.1% of all cardiorenal patients have been diagnosed with acute CRS in India. Acute CRS involves a sudden deterioration of kidney functionalities, referred to as acute kidney injury (AKI) in acute heart failure patients. The pathophysiology of CRS involves hyperactivation of the sympathetic nervous system (SNS) and the renin-angiotensin-aldosterone system (RAAS) following acute myocardial stress. The pathological phenotype of acute CRS is associated with perturbed inflammatory, cellular, and neurohormonal markers in circulation. These complications increase the risk of mortality in clinically diagnosed acute CRS patients, making it a worldwide healthcare burden. Hence, effective diagnosis and early prevention are crucial to prevent the progression of CRS in AHF patients. Present biomarkers, such as serum creatinine (sCr), cystatin C (CysC), glomerular filtration rate (GFR), blood urea nitrogen (BUN), serum and/or urine neutrophil gelatinase-associated lipocalin (NGAL), B-type natriuretic peptide (BNP), and NT-proBNP, are clinically used to diagnose AKI stages in CRS patients but are limitedly sensitive to the early detection of the pathology. Therefore, the need for protein biomarkers is emerging for early intervention in CRS progression. Here, we summarized the cardio-renal nexus in acute CRS, with an emphasis on the present clinicopathological biomarkers and their limitations. The objective of this review is to highlight the need for novel proteomic biomarkers that will curb the burgeoning concern and direct future research trials.
Collapse
Affiliation(s)
- Abhi Dutta
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
| | - Shubham Saha
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
| | - Ajay Bahl
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anupam Mittal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
- BioX Center, Indian Institute of Technology (IIT)-Mandi, Mandi, Himachal Pradesh, India
| |
Collapse
|
11
|
Sinha N, Puri V, Kumar V, Nada R, Rastogi A, Jha V, Puri S. Urinary exosomal miRNA-663a shows variable expression in diabetic kidney disease patients with or without proteinuria. Sci Rep 2023; 13:4516. [PMID: 36934129 PMCID: PMC10024703 DOI: 10.1038/s41598-022-26558-4] [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: 04/28/2022] [Accepted: 12/16/2022] [Indexed: 03/20/2023] Open
Abstract
Heterogeneity in the Diabetic Kidney Disease (DKD) diagnosis makes its rational therapeutics challenging. Although albuminuria characterizes DKD, reports also indicate its prevalence among non-proteinuric. Recent understanding of disease progression has thus inclined the focus on proximal tubular cell damage besides the glomeruli. A non-invasive approach exploiting exosomal miRNA derived from human kidney proximal tubular cell line was, hence, targeted. Upon miRNA profiling, three miRNAs, namely, hsa-miR-155-5p, hsa-miR-28-3p, and hsa-miR-425-5p were found to be significantly upregulated, while hsa-miR-663a was downregulated under diabetic conditions. Among these, hsa-miR-663a downregulation was more pronounced in non-proteinuric than proteinuric DKD subjects and was thus selected for the bioinformatics study. Ingenuity Pathway Analysis (IPA) narrowed on to IL-8 signaling and inflammatory response as the most enriched 'canonical pathway' and 'disease pathway' respectively, during DKD. Further, the putative gene network generated from these enriched pathways revealed experimentally induced diabetes, renal tubular injury, and decreased levels of albumin as part of mapping under 'disease and function'. Genes target predictions and annotations by IPA reiterated miR-663a's role in the pathogenesis of DKD following tubular injury. Overall, the observations might offer an indirect reflection of the underlying mechanism between patients who develop proteinuria and non-proteinuria.
Collapse
Affiliation(s)
- Nisha Sinha
- Centre for Stem Cell Tissue Engineering and Biomedical Excellence, Panjab University, Chandigarh, India
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Veena Puri
- Centre for Systems Biology and Bioinformatics, Panjab University, Chandigarh, India
| | - Vivek Kumar
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritambhra Nada
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashu Rastogi
- Department of Endocrinology and Metabolism, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vivekanand Jha
- The George Institute for Global Health, New Delhi, India.
| | - Sanjeev Puri
- Department of Biotechnology, University Institute of Engineering and Technology (UIET), Panjab University, Chandigarh, India.
| |
Collapse
|
12
|
Wu X, Qian L, Zhao H, Lei W, Liu Y, Xu X, Li J, Yang Z, Wang D, Zhang Y, Zhang Y, Tang R, Yang Y, Tian Y. CXCL12/CXCR4: An amazing challenge and opportunity in the fight against fibrosis. Ageing Res Rev 2023; 83:101809. [PMID: 36442720 DOI: 10.1016/j.arr.2022.101809] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 11/26/2022]
Abstract
Fibrosis is a pathological process caused by abnormal wound healing response, which often leads to excessive deposition of extracellular matrix, distortion of organ architecture, and loss of organ function. Aging is an important risk factor for the development of organ fibrosis. C-X-C receptor 4 (CXCR4) is the predominant chemokine receptor on fibrocytes, C-X-C motif ligand 12 (CXCL12) is the only ligand of CXCR4. Accumulated evidence have confirmed that CXCL12/CXCR4 can be involved in multiple pathological mechanisms in fibrosis, such as inflammation, immunity, epithelial-mesenchymal transition, and angiogenesis. In addition, CXCL12/CXCR4 have also been shown to improve fibrosis levels in many organs including the heart, liver, lung and kidney; thus, they are promising targets for anti-fibrotic therapy. Notably, inhibitors of CXCL12 or CXCR4 also play an important role in various fibrosis-related diseases. In summary, this review systematically summarizes the role of CXCL12/CXCR4 in fibrosis, and this information is of great significance for understanding CXCL12/CXCR4. This will also contribute to the design of further studies related to CXCL12/CXCR4 and fibrosis, and shed light on potential therapies for fibrosis.
Collapse
Affiliation(s)
- Xue Wu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Lu Qian
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, Xi'an, China
| | - Wangrui Lei
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yanqing Liu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Xiaoling Xu
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Jiawen Li
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Zhi Yang
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, Xi'an, China
| | - Du Wang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yuchen Zhang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yan Zhang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ran Tang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yang Yang
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.
| | - Ye Tian
- Deparment of Neurology, Xi'an No.3 Hospital, Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
13
|
Broens B, Duitman JW, Zwezerijnen GJC, Nossent EJ, van der Laken CJ, Voskuyl AE. Novel tracers for molecular imaging of interstitial lung disease: A state of the art review. Autoimmun Rev 2022; 21:103202. [PMID: 36150433 DOI: 10.1016/j.autrev.2022.103202] [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: 08/31/2022] [Accepted: 09/16/2022] [Indexed: 12/14/2022]
Abstract
Interstitial lung disease is an overarching term for a wide range of disorders characterized by inflammation and/or fibrosis in the lungs. Most prevalent forms, among others, include idiopathic pulmonary fibrosis (IPF) and connective tissue disease associated interstitial lung disease (CTD-ILD). Currently, only disease modifying treatment options are available for IPF and progressive fibrotic CTD-ILD, leading to reduction or stabilization in the rate of lung function decline at best. Management of these patients would greatly advance if we identify new strategies to improve (1) early detection of ILD, (2) predicting ILD progression, (3) predicting response to therapy and (4) understanding pathophysiology. Over the last years, positron emission tomography (PET) and single photon emission computed tomography (SPECT) have emerged as promising molecular imaging techniques to improve ILD management. Both are non-invasive diagnostic tools to assess molecular characteristics of an individual patient with the potential to apply personalized treatment. In this review, we encompass the currently available pre-clinical and clinical studies on molecular imaging with PET and SPECT in IPF and CTD-ILD. We provide recommendations for potential future clinical applications of these tracers and directions for future research.
Collapse
Affiliation(s)
- Bo Broens
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Rheumatology and Clinical Immunology, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Infection & Immunity, Inflammatory diseases, Amsterdam, the Netherlands.
| | - Jan-Willem Duitman
- Amsterdam Infection & Immunity, Inflammatory diseases, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Pulmonary Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Experimental Immunology (EXIM), Meibergdreef 9, Amsterdam, the Netherlands.
| | - Gerben J C Zwezerijnen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, the Netherlands.
| | - Esther J Nossent
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences Research Institute, Amsterdam, the Netherlands..
| | - Conny J van der Laken
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Rheumatology and Clinical Immunology, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Infection & Immunity, Inflammatory diseases, Amsterdam, the Netherlands.
| | - Alexandre E Voskuyl
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Rheumatology and Clinical Immunology, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Infection & Immunity, Inflammatory diseases, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Peng W, Kepsch A, Kracht TO, Hasan H, Wijayarathna R, Wahle E, Pleuger C, Bhushan S, Günther S, Kauerhof AC, Planinić A, Fietz D, Schuppe HC, Wygrecka M, Loveland KL, Ježek D, Meinhardt A, Hedger MP, Fijak M. Activin A and CCR2 regulate macrophage function in testicular fibrosis caused by experimental autoimmune orchitis. Cell Mol Life Sci 2022; 79:602. [PMID: 36434305 PMCID: PMC9700630 DOI: 10.1007/s00018-022-04632-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/21/2022] [Accepted: 11/13/2022] [Indexed: 11/27/2022]
Abstract
Experimental autoimmune-orchitis (EAO), a rodent model of chronic testicular inflammation and fibrosis, replicates pathogenic changes seen in some cases of human spermatogenic disturbances. During EAO, increased levels of pro-inflammatory and pro-fibrotic mediators such as TNF, CCL2, and activin A are accompanied by infiltration of leukocytes into the testicular parenchyma. Activin A levels correlate with EAO severity, while elevated CCL2 acting through its receptor CCR2 mediates leukocyte trafficking and recruits macrophages. CCR2 + CXCR4 + macrophages producing extracellular matrix proteins contribute widely to fibrogenesis. Furthermore, testicular macrophages (TMs) play a critical role in organ homeostasis. Therefore, we aimed to investigate the role of the activin A/CCL2-CCR2/macrophage axis in the development of testicular fibrosis. Following EAO induction, we observed lower levels of organ damage, collagen deposition, and leukocyte infiltration (including fibronectin+, collagen I+ and CXCR4+ TMs) in Ccr2-/- mice than in WT mice. Furthermore, levels of Il-10, Ccl2, and the activin A subunit Inhba mRNAs were lower in Ccr2-/- EAO testes. Notably, fibronectin+ TMs were also present in biopsies from patients with impaired spermatogenesis and fibrotic alterations. Overexpression of the activin A antagonist follistatin reduced tissue damage and collagen I+ TM accumulation in WT EAO testes, while treating macrophages with activin A in vitro increased the expression of Ccr2, Fn1, Cxcr4, and Mmp2 and enhanced migration along a CCL2 gradient; these effects were abolished by follistatin. Taken together, our data indicate that CCR2 and activin A promote fibrosis during testicular inflammation by regulating macrophage function. Inhibition of CCR2 or activin A protects against damage progression, offering a promising avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Wei Peng
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Artem Kepsch
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Till O Kracht
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Hiba Hasan
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Rukmali Wijayarathna
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Eva Wahle
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Christiane Pleuger
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Stefan Günther
- ECCPS Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - A Christine Kauerhof
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Ana Planinić
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Hans-Christian Schuppe
- Department of Urology, Paediatric Urology and Andrology, Justus Liebig University of Giessen, Giessen, Germany
| | - Małgorzata Wygrecka
- Center for Infection and Genomics of the Lung, German Center for Lung Research, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Kate L Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Davor Ježek
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Mark P Hedger
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany.
| |
Collapse
|
15
|
Hu S, Liu H, Li Q, Yu Q, Liu X, Xu J, Fu R. Suppressing the activity of CXCR4 down-regulates the expression of renal fibrosis related genes in primary glomerular cells. Transl Pediatr 2022; 11:882-890. [PMID: 35800283 PMCID: PMC9253943 DOI: 10.21037/tp-22-157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/13/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND C-X-C chemokine receptor type 4 (CXCR4) has a certain effect on renal fibrosis, and there are few specific studies in cells. We want to investigate the impact of suppressing CXCR4 activity on the expression of renal fibrosis-related genes in primary glomerular endothelial cells, mesangial cells, and podocytes. METHODS Immunofluorescence assays were used to determine the purity of isolated glomerular endothelial cells, mesangial cells, and podocytes. CXCR4 knockdown cell lines were established by transfecting the short hairpin (sh)RNA against CXCR4. T140 and AMD3100 were used to inhibit the activity of CXCR4. LY294002 was used to inhibit the activity of phosphoinositide 3-kinase (PI3K). The mRNA expression of CXCR4 was determined by real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR). The protein expression of CXCR4, collagen IV, matrix metallopeptidase (MMP)-9, PI3K, Rac1, and vascular cell adhesion protein 1 (VCAM-1) was evaluated by Western blot analysis. RESULTS High purity was observed on isolated primary glomerular endothelial cells and podocytes. However, the purity of isolated mesangial cells was relatively low. The mRNA expression of CXCR4 was significantly suppressed by the transfection of shRNA. Compared to control cells, the expression of CXCR4, collagen IV, MMP-9, PI3K, Rac1, and VCAM-1 were dramatically downregulated in cell lines transfected with shRNA against CXCR4. Furthermore, cell lines treated with T140, AMD3100, or LY294002 also showed downregulated expression of these proteins compared to untreated cells. No significant differences were observed in the protein expression of these proteins between control cells and cells transfected with the shRNA negative control (NC). CONCLUSIONS Suppressing the activity of CXCR4 downregulated the expression of renal fibrosis-related genes in primary glomerular cells, even under a non-inflammatory state.
Collapse
Affiliation(s)
- Shaofan Hu
- Department of Nephrology, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Hong Liu
- Department of Nephrology, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Qun Li
- Department of Nephrology, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Qiang Yu
- Department of Nephrology, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Xiaoqing Liu
- Department of Nephrology, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Jin Xu
- Department of Nephrology, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Rui Fu
- Department of Nephrology, Jiangxi Provincial Children's Hospital, Nanchang, China
| |
Collapse
|
16
|
Tang W, Panja S, Jogdeo CM, Tang S, Ding L, Yu A, Foster KW, Dsouza DL, Chhonker YS, Jensen-Smith H, Jang HS, Boesen EI, Murry DJ, Padanilam B, Oupický D. Modified chitosan for effective renal delivery of siRNA to treat acute kidney injury. Biomaterials 2022; 285:121562. [PMID: 35552115 PMCID: PMC9133205 DOI: 10.1016/j.biomaterials.2022.121562] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/02/2022] [Accepted: 05/01/2022] [Indexed: 11/02/2022]
Abstract
Acute kidney injury (AKI) is characterized by a sudden decrease in renal function and impacts growing number of people worldwide. RNA interference (RNAi) showed potential to treat diseases with no or limited conventional therapies, including AKI. Suitable carriers are needed to protect and selectively deliver RNAi to target cells to fully explore this therapeutic modality. Here, we report on the synthesis of chitosan modified with α-cyclam-p-toluic acid (C-CS) as a novel siRNA carrier for targeted delivery to injured kidneys. We demonstrate that conjugation of the α-cyclam-p-toluic acid to chitosan imparts the C-CS polymer with targeting and antagonistic properties to cells overexpressing chemokine receptor CXCR4. In contrast, the parent α-cyclam-p-toluic acid showed no such properties. Self-assembled C-CS/siRNA nanoparticles rapidly accumulate in the injured kidneys and show long retention in renal tubules. Apoptosis and metabolic and inflammatory pathways induced by p53 are important pathological mechanisms in the development of AKI. Nanoparticles with siRNA against p53 (sip53) were formulated and intravenously injected for attenuation of IRI-AKI. Due to the favorable accumulation in injured kidneys, the treatment with C-CS/sip53 decreased renal injury, extent of renal apoptosis, macrophage and neutrophil infiltration, and improved renal function. Overall, our study suggests that C-CS/siRNA nanoparticles have the potential to effectively accumulate and deliver therapeutic siRNAs to injured kidneys through CXCR4 binding, providing a novel way for AKI therapy.
Collapse
Affiliation(s)
- Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Siyuan Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ao Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kirk W Foster
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Del L Dsouza
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yashpal S Chhonker
- Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Heather Jensen-Smith
- Eppley Institute for Research in Cancer & Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Hee-Seong Jang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Erika I Boesen
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Daryl J Murry
- Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Babu Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
17
|
Losartan ameliorates renal interstitial fibrosis through metabolic pathway and Smurfs-TGF-β/Smad. Biomed Pharmacother 2022; 149:112931. [PMID: 36068784 DOI: 10.1016/j.biopha.2022.112931] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/27/2022] [Accepted: 04/05/2022] [Indexed: 11/22/2022] Open
Abstract
The genesis and development of renal fibrosis involve a variety of pathways closely related to inflammation, cytokines, oxidative stress and metabolic abnormalities. Renal fibrosis is the result of a complex combination of a variety of lesions. Epithelial-mesenchymal transdifferentiation (EMT) of renal tubular epithelial cells is considered the key to renal fibrosis. Losartan is a typical Angiotensin II (ANG II) receptor antagonist and relaxes blood vessels. In this study, we investigated the effects of losartan on Unilateral Ureteral Obstruction (UUO) model mice by studying the changes in the TGF-β/Smad and metabolomics. Male C57BL/6 J mice were intervened with the UUO model and given losartan (10, 20, 30 mg/kg/d) for 28 consecutive days. The results showed that losartan could reduce UUO-induced abnormal serum metabolic spectrum and renal function. It could also improve renal tubular-interstitial injury and fibrosis by reducing tubulointerstitial dilation and collagen deposition. In addition, losartan promoted the expression of Smurf2 and Smurf1, i.e., Smad7 and E3 ubiquitin-linked enzymes, in the nucleus to degrade the type I receptor of TGF-β1 (TβR-I) and P-Smad2/3 to inhibit renal tubular epithelial cells EMT. In summary, these findings indicated that losartan could regulate the TGF-β/Smad and metabolic pathway in UUO model mice through ubiquitination to reduce renal fibrosis.
Collapse
|
18
|
Fayçal CA, Oszwald A, Feilen T, Cosenza-Contreras M, Schilling O, Loustau T, Steinbach F, Schachner H, Langer B, Heeringa P, Rees AJ, Orend G, Kain R. An adapted passive model of anti-MPO dependent crescentic glomerulonephritis reveals matrix dysregulation and is amenable to modulation by CXCR4 inhibition. Matrix Biol 2022; 106:12-33. [PMID: 35032611 DOI: 10.1016/j.matbio.2022.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/18/2021] [Accepted: 01/05/2022] [Indexed: 12/25/2022]
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) are severe inflammatory disorders that often involve focal necrotizing glomerulonephritis (FNGN) and consequent glomerular scarring, interstitial fibrosis, and chronic kidney disease. Robust murine models of scarring in FNGN that may help to further our understanding of deleterious processes are still lacking. Here, we present a murine model of severe FNGN based on combined administration of antibodies against the glomerular basement membrane (GBM) and myeloperoxidase (MPO), and bacterial lipopolysaccharides (LPS), that recapitulates acute injury and was adapted to investigate subsequent glomerular and interstitial scarring. Hematuria without involvement of other organs occurs consistently and rapidly, glomerular necrosis and crescent formation are evident at 12 days, and consequent glomerular and interstitial scarring at 29 days after initial treatment. Using mass-spectrometric proteome analysis, we provide a detailed overview of matrisomal and cellular changes in our model. We observed increased expression of the matrisome including collagens, fibronectin, tenascin-C, in accordance with human AAV as deduced from analysis of gene expression microarrays and tissue staining. Moreover, we observed tissue infiltration by neutrophils, macrophages, T cells and myofibroblasts upon injury. Experimental inhibition of CXCR4 using AMD3100 led to a sustained histological presence of fibrin extravasate, reduced chemokine expression and leukocyte activation, but did not markedly affect ECM composition. Altogether, we demonstrate an adapted FNGN model that enables the study of matrisomal changes both in disease and upon intervention, as exemplified via CXCR4 inhibition.
Collapse
Affiliation(s)
- Chérine Abou Fayçal
- INSERM U1109, The Tumor Microenvironment Laboratory, Strasbourg, France; Université Strasbourg, Hopital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France; These authors (CAF and AO) contributed equally to this work
| | - Andre Oszwald
- Department of Pathology, Medical University of Vienna, Vienna, Austria; These authors (CAF and AO) contributed equally to this work
| | - Tobias Feilen
- Institute of Surgical Pathology, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Miguel Cosenza-Contreras
- Institute of Surgical Pathology, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Thomas Loustau
- INSERM U1109, The Tumor Microenvironment Laboratory, Strasbourg, France; Université Strasbourg, Hopital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Fanny Steinbach
- INSERM U1109, The Tumor Microenvironment Laboratory, Strasbourg, France; Université Strasbourg, Hopital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Helga Schachner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Brigitte Langer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Andrew J Rees
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Gertraud Orend
- INSERM U1109, The Tumor Microenvironment Laboratory, Strasbourg, France; Université Strasbourg, Hopital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| | - Renate Kain
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
19
|
Cao Q, Huang C, Yi H, Gill AJ, Chou A, Foley M, Hosking CG, Lim KK, Triffon CF, Shi Y, Chen XM, Pollock CA. A single domain i-body (AD-114) attenuates renal fibrosis through blockade of CXCR4. JCI Insight 2022; 7:143018. [PMID: 35015734 PMCID: PMC8876455 DOI: 10.1172/jci.insight.143018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
The G protein–coupled CXC chemokine receptor 4 (CXCR4) is a candidate therapeutic target for tissue fibrosis. A fully human single-domain antibody-like scaffold i-body AD-114-PA600 (AD-114) with specific high binding affinity to CXCR4 has been developed. To define its renoprotective role, AD-114 was administrated in a mouse model of renal fibrosis induced by folic acid (FA). Increased extracellular matrix (ECM) accumulation, macrophage infiltration, inflammatory response, TGF-β1 expression, and fibroblast activation were observed in kidneys of mice with FA-induced nephropathy. These markers were normalized or partially reversed by AD-114 treatment. In vitro studies demonstrated AD-114 blocked TGF-β1–induced upregulated expression of ECM, matrix metalloproteinase-2, and downstream p38 mitogen-activated protein kinase (p38 MAPK) and PI3K/AKT/mTOR signaling pathways in a renal proximal tubular cell line. Additionally, these renoprotective effects were validated in a second model of unilateral ureteral obstruction using a second generation of AD-114 (Fc-fused AD-114, also named AD-214). Collectively, these results suggest a renoprotective role of AD-114 as it inhibited the chemotactic function of CXCR4 as well as blocked CXCR4 downstream p38 MAPK and PI3K/AKT/mTOR signaling, which establish a therapeutic strategy for AD-114 targeting CXCR4 to limit renal fibrosis.
Collapse
Affiliation(s)
- Qinghua Cao
- Renal Medicine, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Chunling Huang
- Renal Medicine, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Hao Yi
- Renal Medicine, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Anthony J Gill
- Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, Australia
| | - Angela Chou
- Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, Australia
| | - Michael Foley
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Chris G Hosking
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Kevin K Lim
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Cristina F Triffon
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Ying Shi
- Renal Medicine, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Xin-Ming Chen
- Renal Medicine, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Carol A Pollock
- Renal Medicine, Kolling Institute of Medical Research, University of Sydney, Sydney, Australia
| |
Collapse
|
20
|
Preferential siRNA delivery to injured kidneys for combination treatment of acute kidney injury. J Control Release 2022; 341:300-313. [PMID: 34826532 PMCID: PMC8776616 DOI: 10.1016/j.jconrel.2021.11.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 01/03/2023]
Abstract
Acute kidney injury (AKI) is characterized by a sudden loss of renal function and is associated with high morbidity and mortality. Tumor suppressor p53 and chemokine receptor CXCR4 were both implicated in the AKI pathology. Here, we report on the development and evaluation of polymeric CXCR4 antagonist (PCX) siRNA carrier for selective delivery to injured kidneys in AKI. Our results show that PCX/siRNA nanoparticles (polyplexes) provide protection against cisplatin injury to tubule cells in vitro when both CXCR4 and p53 are inhibited. The polyplexes selectively accumulate and are retained in the injured kidneys in cisplatin and bilateral ischemia reperfusion injury models of AKI. Treating AKI with the combined CXCR4 inhibition and p53 gene silencing with the PCX/sip53 polyplexes improves kidney function and decreases renal damage. Overall, our results suggest that the PCX/sip53 polyplexes have a significant potential to enhance renal accumulation in AKI and deliver therapeutic siRNA.
Collapse
|
21
|
Tanaka M, Kakihara S, Hirabayashi K, Imai A, Toriyama Y, Iesato Y, Sakurai T, Kamiyoshi A, Ichikawa-Shindo Y, Kawate H, Tanaka M, Cui N, Wei Y, Zhao Y, Aruga K, Yamauchi A, Murata T, Shindo T. Adrenomedullin-Receptor Activity-Modifying Protein 2 System Ameliorates Subretinal Fibrosis by Suppressing Epithelial-Mesenchymal Transition in Age-Related Macular Degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:652-668. [PMID: 33385343 DOI: 10.1016/j.ajpath.2020.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 01/06/2023]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment. Anti-vascular endothelial growth factor drugs used to treat AMD carry the risk of inducing subretinal fibrosis. We investigated the use of adrenomedullin (AM), a vasoactive peptide, and its receptor activity-modifying protein 2, RAMP2, which regulate vascular homeostasis and suppress fibrosis. The therapeutic potential of the AM-RAMP2 system was evaluated after laser-induced choroidal neovascularization (LI-CNV), a mouse model of AMD. Neovascular formation, subretinal fibrosis, and macrophage invasion were all enhanced in both AM and RAMP2 knockout mice compared with those in wild-type mice. These pathologic changes were suppressed by intravitreal injection of AM. Comprehensive gene expression analysis of the choroid after LI-CNV with or without AM administration revealed that fibrosis-related molecules, including Tgfb, Cxcr4, Ccn2, and Thbs1, were all down-regulated by AM. In retinal pigment epithelial cells, co-administration of transforming growth factor-β and tumor necrosis factor-α induced epithelial-mesenchymal transition, which was also prevented by AM. Finally, transforming growth factor-β and C-X-C chemokine receptor type 4 (CXCR4) inhibitors eliminated the difference in subretinal fibrosis between RAMP2 knockout and wild-type mice. These findings suggest the AM-RAMP2 system suppresses subretinal fibrosis in LI-CNV by suppressing epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Masaaki Tanaka
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Shinji Kakihara
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | | | - Akira Imai
- Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Yuichi Toriyama
- Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Yasuhiro Iesato
- Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Akiko Kamiyoshi
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Yuka Ichikawa-Shindo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hisaka Kawate
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Megumu Tanaka
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Nanqi Cui
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yangxuan Wei
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yunlu Zhao
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kohsuke Aruga
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Akihiro Yamauchi
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Toshinori Murata
- Department of Ophthalmology, Shinshu University School of Medicine, Japan
| | - Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University School of Medicine, Matsumoto, Japan; Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan.
| |
Collapse
|
22
|
Makitani K, Ogo N, Asai A. STX-0119, a novel STAT3 dimerization inhibitor, prevents fibrotic gene expression in a mouse model of kidney fibrosis by regulating Cxcr4 and Ccr1 expression. Physiol Rep 2020; 8:e14627. [PMID: 33112058 PMCID: PMC7592413 DOI: 10.14814/phy2.14627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/30/2022] Open
Abstract
Kidney fibrosis is a histological hallmark of chronic kidney disease (CKD) and is believed to be involved in the progression of CKD. Therefore, inhibition of kidney fibrosis is a potential strategy for slowing CKD progression. Signal transducer and activator of transcription 3 (STAT3) is a transcription factor that is activated by interleukin-6 and is reported to be involved in fibrosis. Previously, S3I-201, an inhibitor of STAT3 phosphorylation, was shown to inhibit renal fibrosis in a mouse model, but its mechanism was not clarified completely. In this study, we investigated whether STX-0119, a new inhibitor of STAT3 dimerization, suppressed kidney fibrotic gene expression using a mouse model of kidney fibrosis and examined the underlying mechanisms. Kidney fibrosis was induced by unilateral ureteral obstruction (UUO), which was accompanied by upregulation of STAT3 target genes. STX-0119 administration suppressed the expression of fibrotic genes in UUO kidneys without affecting STAT3 phosphorylation. STX-0119 decreased Cxcr4 mRNA in cultured rat kidney fibroblasts and Ccr1 mRNA in blood cells from UUO mice, both of which are reported to be involved in the progression of kidney fibrosis. These results suggest that STX-0119 inhibits fibrotic gene expression in kidney by suppressing Cxcr4 and Ccr1 expression. This is the first report to indicate a part of the mechanism of the antifibrotic effects of a STAT3 inhibitor and suggests that STX-0119 may be a lead compound for the treatment of kidney fibrosis.
Collapse
Affiliation(s)
- Kouki Makitani
- Center for Drug DiscoveryGraduate School of Pharmaceutical SciencesUniversity of ShizuokaShizuokaJapan
| | - Naohisa Ogo
- Center for Drug DiscoveryGraduate School of Pharmaceutical SciencesUniversity of ShizuokaShizuokaJapan
| | - Akira Asai
- Center for Drug DiscoveryGraduate School of Pharmaceutical SciencesUniversity of ShizuokaShizuokaJapan
| |
Collapse
|
23
|
Szczepańska M, Sędek Ł, Bulsa J, Mazur B, Zwolińska D, Tkaczyk M, Karpe J, Szczepański T. Chemokine receptors on peripheral blood T lymphocytes in children on peritoneal dialysis. Perit Dial Int 2020; 41:194-201. [PMID: 32869708 DOI: 10.1177/0896860820951292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Immune cell dysfunction is listed among complications resulting from chronic kidney disease (CKD). It could be associated with T-cells, which play a role in the lymphocytic migration and infiltration. However, the data on chemokine receptors expression on T-cells in patients with CKD particularly treated with peritoneal dialysis (PD) are still limited. METHODS The study aimed at multiparameter flow-cytometric analysis of the absolute numbers and percentage of T-cell subsets with surface chemokine receptors (CCR4, CCR5, CCR7, CXCR3, and CXCR4) or receptors' combinations in 47 children treated with PD. RESULTS We found lower absolute numbers of total T lymphocytes, lymphocytes with surface CCR5, CXCR4+CCR5, CXCR3+CCR5 antigens and T-cells with CCR4, CCR4+CD4, CXCR3, CXCR3+CD4, and CD8 receptors. Lymphocytes T with CD4, CCR7, CD28+CCR7, CXCR3+CD8 antigens showed higher percentage in children on PD as compared to healthy children and opposite percentage values of CCR4+, CCR4+CD4+, CXCR3+ T lymphocytes were diminished. Mean fluorescent intensity for CCR7+, CCR7+CD45RO+, CCR7+CD28+, CXCR4+CD4+, CCR5+CD4+, CCR4+, CCR4+CD4+ T-cells was lower in the PD group than in healthy children. The analysis of correlation between T lymphocyte subpopulations with chemokine receptors and other parameters revealed positive correlation of CCR7+ and CCR7+CD28+ T-cells and weekly creatinine clearance, negative correlation between the percentage of CD45RO+CCR7 antigen positive T-cells and KT/Vurea. SUMMARY In conclusion, we could not confirm the phenomenon of earlier senescence of T-cells in children with CKD on PD treatment. This still requires further investigation. The higher percentage of T-cells with CCR7 surface receptor could be responsible for the increase of proliferation activity in this group of children.
Collapse
Affiliation(s)
- Maria Szczepańska
- Department of Pediatrics, 49613Faculty of Medical Sciences in Zabrze, SUM in Katowice, Poland
| | - Łukasz Sędek
- Department of Microbiology and Immunology, 49613Faculty of Medical Sciences in Zabrze, SUM in Katowice, Poland
| | - Joanna Bulsa
- Department of Pediatric Hematology and Oncology, 49613Faculty of Medical Sciences in Zabrze, SUM in Katowice, Poland
| | - Bogdan Mazur
- Department of Microbiology and Immunology, 49613Faculty of Medical Sciences in Zabrze, SUM in Katowice, Poland
| | - Danuta Zwolińska
- Department of Pediatric Nephrology, Wrocław Medical University, Poland
| | - Marcin Tkaczyk
- Department of Pediatrics, Immunology and Nephrology, 49602Polish Mother's Memorial Hospital Research Institute of Lodz, Poland
| | - Jacek Karpe
- Department of Anesthesiology and Intensive Care, 49613Faculty of Medical Sciences in Zabrze, SUM in Katowice, Poland
| | - Tomasz Szczepański
- Department of Pediatric Hematology and Oncology, 49613Faculty of Medical Sciences in Zabrze, SUM in Katowice, Poland
| |
Collapse
|
24
|
Derlin T, Jaeger B, Jonigk D, Apel RM, Freise J, Shin HO, Weiberg D, Warnecke G, Ross TL, Wester HJ, Seeliger B, Welte T, Bengel FM, Prasse A. Clinical Molecular Imaging of Pulmonary CXCR4 Expression to Predict Outcome of Pirfenidone Treatment in Idiopathic Pulmonary Fibrosis. Chest 2020; 159:1094-1106. [PMID: 32822674 DOI: 10.1016/j.chest.2020.08.2043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/03/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive disease for which two antifibrotic drugs recently were approved. However, an unmet need exists to predict responses to antifibrotic treatment, such as pirfenidone. Recent data suggest that upregulated expression of CXCR4 is indicative of outcomes in IPF. RESEARCH QUESTION Can quantitative, molecular imaging of pulmonary CXCR4 expression as a biomarker for disease activity predict response to the targeted treatment pirfenidone and prognosis in patients with IPF? STUDY DESIGN AND METHODS CXCR4 expression was analyzed by immunohistochemistry examination of lung tissues and reverse-transcriptase polymerase chain reaction analysis of BAL. PET-CT scanning with the specific CXCR4 ligand 68Ga-pentixafor was performed in 28 IPF patients and compared with baseline clinical characteristics. In 16 patients, a follow-up scan was obtained 6 to 12 weeks after initiation of treatment with pirfenidone. Patients were followed up in our outpatient clinic for ≥ 12 months. RESULTS Immunohistochemistry analysis showed high CXCR4 staining of epithelial cells and macrophages in areas with vast fibrotic remodeling. Targeted PET scanning revealed CXCR4 upregulation in fibrotic areas of the lungs, particularly in zones with subpleural honeycombing. Baseline CXCR4 signal demonstrated a significant correlation with Gender Age Physiology stage (r = 0.44; P = .02) and with high-resolution CT scan score (r = 0.38; P = .04). Early changes in CXCR4 signal after initiation of pirfenidone treatment correlated with the long-term course of FVC after 12 months (r = -0.75; P = .0008). Moreover, patients with a high pulmonary CXCR4 signal on follow-up PET scan after 6 weeks into treatment demonstrated a statistically significant worse outcome at 12 months (P = .002). In multiple regression analysis, pulmonary CXCR4 signal on follow-up PET scan emerged as the only independent predictor of long-term outcome (P = .0226). INTERPRETATION CXCR4-targeted PET imaging identified disease activity and predicted outcome of IPF patients treated with pirfenidone. It may serve as a future biomarker for personalized guidance of antifibrotic treatment.
Collapse
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Benedikt Jaeger
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Rosa M Apel
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Julia Freise
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Hoen-Oh Shin
- Institute of Radiology, Hannover Medical School, Hannover, Germany
| | - Desiree Weiberg
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Gregor Warnecke
- Department of Heart, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Hans-Jürgen Wester
- Institute of Radiopharmaceutical Chemistry, Technical University Munich, Garching, Germany
| | - Benjamin Seeliger
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Tobias Welte
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Antje Prasse
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; DZL-BREATH, Hannover, Germany.
| |
Collapse
|
25
|
Zhang S, Breidenbach JD, Khalaf FK, Dube PR, Mohammed CJ, Lad A, Stepkowski S, Hinds TD, Kumarasamy S, Kleinhenz A, Tian J, Malhotra D, Kennedy DJ, Cooper CJ, Haller ST. Renal Fibrosis Is Significantly Attenuated Following Targeted Disruption of Cd40 in Experimental Renal Ischemia. J Am Heart Assoc 2020; 9:e014072. [PMID: 32200719 PMCID: PMC7428653 DOI: 10.1161/jaha.119.014072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Renal artery stenosis is a common cause of renal ischemia, contributing to the development of chronic kidney disease. To investigate the role of local CD40 expression in renal artery stenosis, Goldblatt 2‐kidney 1‐clip surgery was performed on hypertensive Dahl salt‐sensitive rats (S rats) and genetically modified S rats in which CD40 function is abolished (Cd40mutant). Methods and Results Four weeks following the 2‐kidney 1‐clip procedure, Cd40mutant rats demonstrated significantly reduced blood pressure and renal fibrosis in the ischemic kidneys compared with S rat controls. Similarly, disruption of Cd40 resulted in reduced 24‐hour urinary protein excretion in Cd40mutant rats versus S rat controls (46.2±1.9 versus 118.4±5.3 mg/24 h; P<0.01), as well as protection from oxidative stress, as indicated by increased paraoxonase activity in Cd40mutant rats versus S rat controls (P<0.01). Ischemic kidneys from Cd40mutant rats demonstrated a significant decrease in gene expression of the profibrotic mediator, plasminogen activator inhibitor‐1 (P<0.05), and the proinflammatory mediators, C‐C motif chemokine ligand 19 (P<0.01), C‐X‐C Motif Chemokine Ligand 9 (P<0.01), and interleukin‐6 receptor (P<0.001), compared with S rat ischemic kidneys, as assessed by quantitative PCR assay. Reciprocal renal transplantation documented that CD40 exclusively expressed in the kidney contributes to ischemia‐induced renal fibrosis. Furthermore, human CD40‐knockout proximal tubule epithelial cells suggested that suppression of CD40 signaling significantly inhibited expression of proinflammatory and ‐fibrotic genes. Conclusions Taken together, our data suggest that activation of CD40 induces a significant proinflammatory and ‐fibrotic response and represents an attractive therapeutic target for treatment of ischemic renal disease.
Collapse
Affiliation(s)
- Shungang Zhang
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Joshua D. Breidenbach
- Department of Medical Microbiology and ImmunologyUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Fatimah K. Khalaf
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Prabhatchandra R. Dube
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Chrysan J. Mohammed
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Apurva Lad
- Department of Medical Microbiology and ImmunologyUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Stanislaw Stepkowski
- Department of Medical Microbiology and ImmunologyUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Terry D. Hinds
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Sivarajan Kumarasamy
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Andrew Kleinhenz
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Jiang Tian
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Deepak Malhotra
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - David J. Kennedy
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Christopher J. Cooper
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| | - Steven T. Haller
- Department of MedicineUniversity of Toledo College of Medicine and Life SciencesToledoOH
| |
Collapse
|
26
|
Liu Y, Feng Q, Miao J, Wu Q, Zhou S, Shen W, Feng Y, Hou FF, Liu Y, Zhou L. C-X-C motif chemokine receptor 4 aggravates renal fibrosis through activating JAK/STAT/GSK3β/β-catenin pathway. J Cell Mol Med 2020; 24:3837-3855. [PMID: 32119183 PMCID: PMC7171406 DOI: 10.1111/jcmm.14973] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/18/2019] [Accepted: 12/24/2019] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) has a high prevalence worldwide. Renal fibrosis is the common pathological feature in various types of CKD. However, the underlying mechanisms are not determined. Here, we adopted different CKD mouse models and cultured human proximal tubular cell line (HKC-8) to examine the expression of C-X-C motif chemokine receptor 4 (CXCR4) and β-catenin signalling, as well as their relationship in renal fibrosis. In CKD mice and humans with a variety of nephropathies, CXCR4 was dramatically up-regulated in tubules, with a concomitant activation of β-catenin. CXCR4 expression level was positively correlated with the expression of β-catenin target MMP-7. AMD3100, a CXCR4 receptor blocker, and gene knockdown of CXCR4 significantly inhibited the activation of JAK/STAT and β-catenin signalling, protected against tubular injury and renal fibrosis. CXCR4-induced renal fibrosis was inhibited by treatment with ICG-001, an inhibitor of β-catenin signalling. In HKC-8 cells, overexpression of CXCR4 induced activation of β-catenin and deteriorated cell injury. These effects were inhibited by ICG-001. Stromal cell-derived factor (SDF)-1α, the ligand of CXCR4, stimulated the activation of JAK2/STAT3 and JAK3/STAT6 signalling in HKC-8 cells. Overexpression of STAT3 or STAT6 decreased the abundance of GSK3β mRNA. Silencing of STAT3 or STAT6 significantly blocked SDF-1α-induced activation of β-catenin and fibrotic lesions. These results uncover a novel mechanistic linkage between CXCR4 and β-catenin activation in renal fibrosis in association with JAK/STAT/GSK3β pathway. Our studies also suggest that targeted inhibition of CXCR4 may provide better therapeutic effects on renal fibrosis by inhibiting multiple downstream signalling cascades.
Collapse
Affiliation(s)
- Yahong Liu
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Division of Nephrology, The Second Affiliated Hospital of Xingtai Medical College, Xingtai, China
| | - Qijian Feng
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinhua Miao
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qinyu Wu
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shan Zhou
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Shen
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Terpstra ML, Remmerswaal EBM, van Aalderen MC, Wever JJ, Sinnige MJ, van der Bom-Baylon ND, Bemelman FJ, Geerlings SE. Circulating mucosal-associated invariant T cells in subjects with recurrent urinary tract infections are functionally impaired. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:80-92. [PMID: 32032475 PMCID: PMC7016840 DOI: 10.1002/iid3.287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022]
Abstract
Background Urinary tract infection recurrence is common, particularly in women and immunocompromised patients, such as renal transplant recipients (RTRs). Mucosal‐associated invariant T (MAIT) cells play a role in the antibacterial response by recognizing bacterial riboflavin metabolites produced by bacteria such as Escherichia coli. Here, we investigated whether MAIT cells are involved in the pathogenesis of recurrent urinary tract infections (RUTIs). Methods Using multichannel flow cytometry, we characterized the MAIT cell phenotype and function in blood from immunocompetent adults with (n = 13) and without RUTIs (n = 10) and in RTRs with (n = 9) and without RUTIs (n = 10). Results There were no differences in the numbers of MAIT cells between the study groups. MAIT cells in patients with RUTI expressed T‐bet more often than those in controls. MAIT cells from immunocompetent RUTI participants required more antigen‐presenting cells coincubated with E. coli to evoke a similar cytokine and degranulation response than those from controls. This effect was absent in the RTR with RUTI vs RTR control groups, where the overall percentage of MAIT cells that responded to stimulation was already reduced. Conclusion Circulating MAIT cells in immunocompetent individuals with RUTIs respond to bacterial stimuli with reduced efficacy, which suggests that they are involved in the pathogenesis of RUTIs.
Collapse
Affiliation(s)
- Matty L Terpstra
- Division of Nephrology, Department of Internal Medicine, Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Michiel C van Aalderen
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joyce J Wever
- Division of Nephrology, Department of Internal Medicine, Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marjan J Sinnige
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nelly D van der Bom-Baylon
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frederike J Bemelman
- Division of Nephrology, Department of Internal Medicine, Renal Transplant Unit, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Suzanne E Geerlings
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
28
|
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
|
29
|
Cao J, Zhu W, Yu D, Pan L, Zhong L, Xiao Y, Gao Y, Jiao Y, Zhang Q, Ji J, Yang H, Zhang S, Cao J. The Involvement of SDF-1α/CXCR4 Axis in Radiation-Induced Acute Injury and Fibrosis of Skin. Radiat Res 2019; 192:410-421. [PMID: 31390312 DOI: 10.1667/rr15384.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced acute skin injury and consequent fibrosis are common complications of cancer radiotherapy and radiation accidents. Stromal cell-derived factor-1α (SDF-1α) and its receptor, CXC chemokine receptor 4 (CXCR4) have been shown to be involved in multiple cellular events. However, the role of SDF-1α/CXCR4 axis in radiation-induced acute injury and fibrosis of skin has not been reported. In this study, we found that the expression of SDF-1α and CXCR4 was significantly increased in irradiated skin tissues of humans, monkeys and rats, compared to their nonirradiated counterparts. Mice with keratinocyte-specific ablation of CXCR4 showed less severe skin damage than wild-type mice after receiving a 35 Gy dose of radiation. Consistently, subcutaneous injection of AMD3100, an FDA approved SDF-1α/CXCR4 inhibitor, attenuated skin injury and fibrosis induced by exposure to radiation in a rat model. Mechanically, the SDF-1α/CXCR4 axis promotes pro-fibrotic TGF-b/Smad signaling through the PI3K-MAPK signaling cascade in human keratinocyte HaCaT cells and skin fibroblast WS1 cells. AMD3100 inhibited Smad2 nuclear translocation and transcriptional activity of Smad2/3 induced by radiation, which suppressed the pro-fibrotic TGF-b/Smad signaling pathway activated by exposure. Taken together, these findings demonstrate the involvement of SDF-1α/CXCR4 axis in radiation-induced acute injury and fibrosis of skin, and indicate that AMD3100 would be an effective countermeasure against these diseases.
Collapse
Affiliation(s)
- Jinming Cao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wei Zhu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Daojiang Yu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.,Departments of Plastic Surgery
| | - Lu Pan
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Li Zhong
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yuji Xiao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yiying Gao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yang Jiao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Qi Zhang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jiang Ji
- Departments of Dermatology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Hongying Yang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Shuyu Zhang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.,Department of Oncology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213100, China.,Department of Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu 610051, China
| | - Jianping Cao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
30
|
Jackson EK, Mi E, Ritov VB, Gillespie DG. Extracellular Ubiquitin(1-76) and Ubiquitin(1-74) Regulate Cardiac Fibroblast Proliferation. Hypertension 2019; 72:909-917. [PMID: 30354710 DOI: 10.1161/hypertensionaha.118.11666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SDF-1α (stromal cell-derived factor-1α) is a CXCR4-receptor agonist and DPP4 (dipeptidyl peptidase 4) substrate. SDF-1α, particularly when combined with sitagliptin to block the metabolism of SDF-1α by DPP4, stimulates proliferation of cardiac fibroblasts via the CXCR4 receptor; this effect is greater in cells from spontaneously hypertensive rats versus Wistar-Kyoto normotensive rats. Emerging evidence indicates that ubiquitin(1-76) exists in plasma and is a potent CXCR4-receptor agonist. Therefore, we hypothesized that ubiquitin(1-76), similar to SDF-1α, should increase proliferation of cardiac fibroblasts. Contrary to our working hypothesis, ubiquitin(1-76) did not stimulate cardiac fibroblast proliferation, yet unexpectedly antagonized the proproliferative effects of SDF-1α combined with sitagliptin. In this regard, ubiquitin(1-76) was more potent in spontaneously hypertensive versus Wistar-Kyoto cells. In the presence of 6bk (selective inhibitor of insulin-degrading enzyme [IDE]; an enzyme known to convert ubiquitin(1-76) to ubiquitin(1-74)), ubiquitin(1-76) no longer antagonized the proproliferative effects of SDF-1α/sitagliptin. Ubiquitin(1-74) also antagonized the proproliferative effects of SDF-1α/sitagliptin, and this effect of ubiquitin(1-74) was not blocked by 6bk and was >10-fold more potent compared with ubiquitin(1-76). Neither ubiquitin(1-76) nor ubiquitin(1-74) inhibited the proproliferative effects of the non-CXCR4 receptor agonist neuropeptide Y (activates Y1 receptors). Cardiac fibroblasts expressed IDE mRNA, protein, and activity and converted ubiquitin(1-76) to ubiquitin(1-74). Spontaneously hypertensive fibroblasts expressed greater IDE activity. Extracellular ubiquitin(1-76) blocks the proproliferative effects of SDF-1α/sitagliptin via its conversion by IDE to ubiquitin(1-74), a potent CXCR4 antagonist. Thus, IDE inhibitors, particularly when combined with DPP4 inhibitors or hypertension, could increase the risk of cardiac fibrosis.
Collapse
Affiliation(s)
- Edwin K Jackson
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| | - Eric Mi
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| | - Vladimir B Ritov
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| | - Delbert G Gillespie
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA
| |
Collapse
|
31
|
Narváez A, Guiteras R, Sola A, Manonelles A, Morote J, Torras J, Grinyó JM, Cruzado JM. siRNA-silencing of CD40 attenuates unilateral ureteral obstruction-induced kidney injury in mice. PLoS One 2019; 14:e0215232. [PMID: 30978213 PMCID: PMC6461348 DOI: 10.1371/journal.pone.0215232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/28/2019] [Indexed: 12/23/2022] Open
Abstract
Background The costimulatory CD40-CD40L pathway plays a role in kidney inflammation. We have previously reported that renal CD40 upregulation precedes cellular interstitial infiltrate and fibrosis in the unilateral ureteral obstruction (UUO) model. Here we sought to evaluate whether the administration of siRNA-CD40 has a therapeutic effect in a reversible unilateral ureteral obstruction (D-UUO) mice model. Methods Eight week-old C57BL6J male mice were divided into four groups: Vehicle (Phosphate-buffered saline) (n = 8); siRNA SC (non-specific siRNA) (n = 6); siRNA-CD40 (n = 8) and WT (wild type) (n = 6) mice. UUO was performed with a microvascular clamp. At day 3 after surgery, the ureteral clamp was removed and nephrectomy of the contralateral kidney was performed. Immediately, PBS, siRNA SC (50μg) or siRNA-CD40 (50μg) was administrated via the tail vein. Mice were killed 48h hours after the siRNA or saline administration. Wild type (WT) mice were used as controls. Blood samples were collected for measuring creatinine and blood urea nitrogen (BUN). Histology and kidney mRNA expression were performed. Results The administration of siRNA-CD40 reduced significantly the severity of acute renal failure associated with UUO. Pathologic analysis showed reduction of tubular dilation, interstitial fibrosis, F4/80 macrophage and CD3 (T cell) infiltration in animals treated with siRNA-CD40. Furthermore, kidney mRNA gene expression analysis showed significantly lower levels of macrophage markers (F4/80 and Mannose receptor), fibrosis matrix proteins (Fibronectin, MMP-9, Collagen IV and α-SMA), pro-inflammatory cytokines (iNOS and MCP-1) and the pro-fibrotic molecule TGF-β1 in siRNA-CD40. Conclusions The administration of siRNA-CD40 therapy reduces the severity of the acute kidney injury induced by obstructive uropathy and promotes kidney repair. This strategy seems suitable to be tested in humans.
Collapse
Affiliation(s)
- Alonso Narváez
- Experimental Nephrology, Department of Ciències Clíniques, Universitat de Barcelona, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Urology, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Roser Guiteras
- Experimental Nephrology, Department of Ciències Clíniques, Universitat de Barcelona, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Anna Sola
- Experimental Nephrology, Department of Ciències Clíniques, Universitat de Barcelona, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Anna Manonelles
- Experimental Nephrology, Department of Ciències Clíniques, Universitat de Barcelona, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Nephrology, Bellvitge University Hospital, Barcelona, Spain
| | - Juan Morote
- Department of Urology, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Juan Torras
- Experimental Nephrology, Department of Ciències Clíniques, Universitat de Barcelona, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M. Grinyó
- Experimental Nephrology, Department of Ciències Clíniques, Universitat de Barcelona, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M. Cruzado
- Experimental Nephrology, Department of Ciències Clíniques, Universitat de Barcelona, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Nephrology, Bellvitge University Hospital, Barcelona, Spain
- * E-mail:
| |
Collapse
|
32
|
Sai L, Yu G, Bo C, Zhang Y, Du Z, Li C, Zhang Z, Jia Q, Shao H, Peng C. Profiling long non-coding RNA changes in silica-induced pulmonary fibrosis in rat. Toxicol Lett 2019; 310:7-13. [PMID: 30978436 DOI: 10.1016/j.toxlet.2019.04.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/23/2019] [Accepted: 04/01/2019] [Indexed: 12/15/2022]
Abstract
Silicosis is a kind of chronic and incurable lung fibrotic disease with pathogenesis and molecular mechanisms largely unknown. Mounting evidence suggests that long non-coding RNAs (lncRNAs) are involved in the pathogenesis of silicosis. However, how many lncRNAs involved in the pulmonary fibrosis remains to be elucidated. In this study, Wistar rats were exposed to silicon dioxide by an improved tracheal intubation method. Rats in the control group were treated with normal saline solution. Results showed that 28 days after exposure, there were significant differences in body weight and lung coefficient of rats treated with silica compared with control rats. The formation of lung fibrosis in silica-induced rats was confirmed by histologic examination. We then investigated the lncRNAs expression changes in lung tissues of silica-exposed rats and compared that with the rats in the control group using microarray. The results indicated that silica exposure leads to altered expression profile in 682 lncRNAs (300 upregulated and 382 downregulated). Seventy-three ceRNA pairs were acquired by predicted analysis. Kyoto Encyclopedia of Genes and Genomes pathway and Gene Ontology analyses were used to predict the biological pathway and functional classification of lncRNAs. The results showed that silica exposure affected 13 lncRNAs pathways. The functional classification mainly involved in protein binding, cell shape and extracellular exosome. This study indicated that alteration of lncRNAs may play a role in silica-induced pulmonary fibrosis through regulation of expressions of functional genes in lungs of rat. Our results provide more insights into the mechanism of silicosis.
Collapse
Affiliation(s)
- Linlin Sai
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China
| | - Gongchang Yu
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China
| | - Cunxiang Bo
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China
| | - Yu Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China
| | - Zhongjun Du
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China
| | - Zhenling Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China.
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China.
| | - Cheng Peng
- Shandong Academy of Occupational Health and Occupational Medicine, Ji'nan, Shandong, China; The University of Queensland, Queensland Alliance for Environmental Health Sciences (QAEHS), Brisbane, Queensland, Australia
| |
Collapse
|
33
|
Tang H, Xu Y, Zhang Z, Zeng S, Dong W, Jiao W, Hu X. SDF‑1/CXCR4 induces epithelial‑mesenchymal transition through activation of the Wnt/β‑catenin signaling pathway in rat chronic allograft nephropathy. Mol Med Rep 2019; 19:3696-3706. [PMID: 30896799 PMCID: PMC6470988 DOI: 10.3892/mmr.2019.10045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/06/2019] [Indexed: 12/03/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been demonstrated to serve a crucial role in the progression of interstitial fibrosis, which is one of the principal pathological features of chronic allograft nephropathy (CAN). However, to the best of our knowledge, the mechanisms of EMT in CAN have not been investigated. In the present study, the effect of stromal cell-derived factor 1 (SDF-1) and the Wnt signaling pathway on the progression of EMT following kidney transplantation was investigated. The CAN model was established using Fisher 344 and Lewis rats, treated with low-dose cyclosporine with or without AMD3100. CAN was confirmed by the pathological alterations and chronic allograft damage index scoring, and EMT was confirmed by western blotting and reverse transcription-quantitative polymerase chain reaction. In the AMD3100 group, there were lower expression levels of α-SMA and higher expression levels of E-cadherin, which indicated that CAN and EMT were ameliorated by AMD3100. The kidney tissue was analyzed using an mRNA + long noncoding (lnc)RNA microarray. A total of 506 mRNAs and 404 lncRNAs were demonstrated to be significantly differentially expressed between the two groups, which revealed the involvement of SDF-1/CXC chemokine receptor 4 (CXCR4) and the Wnt pathway. SDF-1 was demonstrated to induce EMT in vitro through the upregulation of α-SMA, downregulation of E-cadherin and the wound healing assay, and in the rat renal tubular epithelial cells via the nuclear accumulation of β-catenin, which were all inhibited by either AMD3100 or DKK-1. CXXC finger protein 5 (CXXC5), a negative regulator of the Wnt pathway, was downregulated following treatment with SDF-1, which was inhibited by AMD3100 but not by DKK-1. Thus, CXXC5 may be a regulator downstream of SDF-1/CXCR4 in EMT. In conclusion, SDF-1/CXCR4 induces EMT of renal tubular epithelial cells with the involvement of the Wnt pathway, which may be a novel mechanism and therapeutic target in kidney allograft fibrosis of rats.
Collapse
Affiliation(s)
- Hao Tang
- Urology Institute of Capital Medical University, Department of Urology, Capital Medical University Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Yue Xu
- Urology Institute of Capital Medical University, Department of Urology, Capital Medical University Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Zijian Zhang
- Urology Institute of Capital Medical University, Department of Urology, Capital Medical University Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Song Zeng
- Urology Institute of Capital Medical University, Department of Urology, Capital Medical University Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Wenbo Dong
- Urology Institute of Capital Medical University, Department of Urology, Capital Medical University Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Wenjiao Jiao
- Urology Institute of Capital Medical University, Department of Urology, Capital Medical University Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| | - Xiaopeng Hu
- Urology Institute of Capital Medical University, Department of Urology, Capital Medical University Beijing Chaoyang Hospital, Beijing 100020, P.R. China
| |
Collapse
|
34
|
Role of the high-affinity leukotriene B4 receptor signaling in fibrosis after unilateral ureteral obstruction in mice. PLoS One 2019; 14:e0202842. [PMID: 30818366 PMCID: PMC6394974 DOI: 10.1371/journal.pone.0202842] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 02/10/2019] [Indexed: 12/28/2022] Open
Abstract
Leukotriene B4 (LTB4) is a lipid mediator that acts as a potent chemoattractant for inflammatory leukocytes. Kidney fibrosis is caused by migrating inflammatory cells and kidney-resident cells. Here, we examined the role of the high-affinity LTB4 receptor BLT1 during development of kidney fibrosis induced by unilateral ureteral obstruction (UUO) in wild-type (WT) mice and BLT1 knockout (BLT1-/-) mice. We found elevated expression of 5-lipoxygenase (5-LOX), which generates LTB4, in the renal tubules of UUO kidneys from WT mice and BLT1-/- mice. Accumulation of immunoreactive type I collagen in WT UUO kidneys increased over time; however, the increase was less prominent in BLT1-/- UUO kidneys. Accumulation of S100A4-positive fibroblasts increased temporally in WT UUO kidneys, but was again less pronounced in-BLT1-/- UUO kidneys. The same was true of mRNA encoding transforming growth factor-β (TGF)-β and fibroblast growth factor (FGF)-2. Finally, accumulation of F4/80-positive macrophages, which secrete TGF-β, increased temporally in WT UUO and BLT1-/- UUO kidneys, but to a lesser extent in the latter. Following LTB4 stimulation in vitro, macrophages showed increased expression of mRNA encoding TGF-β/FGF-2 and Col1a1, whereas L929 fibroblasts showed increased expression of mRNA encoding α smooth muscle actin (SMA). Bone marrow (BM) transplantation studies revealed that the area positive for type I collagen was significantly smaller in BLT1-/—BM→WT than in WT-BM→WT. Thus, LTB4-BLT1 signaling plays a critical role in fibrosis in UUO kidneys by increasing accumulation of macrophages and fibroblasts. Therefore, blocking BLT1 may prevent renal fibrosis.
Collapse
|
35
|
Troxerutin Protects Kidney Tissue against BDE-47-Induced Inflammatory Damage through CXCR4-TXNIP/NLRP3 Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9865495. [PMID: 29849929 PMCID: PMC5932985 DOI: 10.1155/2018/9865495] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/07/2017] [Indexed: 12/11/2022]
Abstract
2,2′,4,4′-Tetrabromodiphenyl ether (BDE-47) induces oxidative stress in kidney cells, but the underlying mechanism remains poorly understood. Troxerutin, a natural flavonoid, has potential antioxidant and anti-inflammatory efficacy. In this study, we assessed the effect of troxerutin on kidney damage caused by BDE-47 and investigated the underlying mechanism. The results showed troxerutin reduced reactive oxygen species (ROS) level and urine albumin-to-creatinine ratio (ACR), decreased the activities of inflammatory factors including cyclooxygenase-2 (COX-2), induced nitric oxide synthase (iNOS) and nuclear factor kappa B (NF-κB) in the kidney tissues of BDE-47-treated mice. Furthermore, troxerutin significantly weakened the expression of kidney NLRP3 inflammasome containing NLRP3, ASC, and caspase-1, contributing to the decline of IL-1β. Additionally, troxerutin inhibited the increased protein level of stromal-derived factor-1(SDF-1), C-X-C chemokine ligand 12 receptor 4 (CXCR4), and thioredoxin interaction protein (TXNIP) caused by BDE-47. Specifically, the immunoprecipitation assay indicated that there was a direct interaction between CXCR4 and TXNIP. CXCR4 siRNA and TXNIP siRNA also decreased the inflammatory damage, which was similar to the action of troxerutin. Our data demonstrated that troxerutin regulated the inflammatory lesions via CXCR4-TXNIP/NLRP3 inflammasome in the kidney of mice induced by BDE-47.
Collapse
|
36
|
Habiel DM, Espindola MS, Coelho AL, Hogaboam CM. Modeling Idiopathic Pulmonary Fibrosis in Humanized Severe Combined Immunodeficient Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:891-903. [PMID: 29378172 PMCID: PMC5954978 DOI: 10.1016/j.ajpath.2017.12.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 12/11/2017] [Accepted: 12/28/2017] [Indexed: 12/17/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fibrotic lung disease of unknown etiopathogenesis with limited therapeutic options. IPF is characterized by an abundance of fibroblasts and loss of epithelial progenitors, which cumulates in unrelenting fibrotic lung remodeling and loss of normal oxygenation. IPF has been challenging to model in rodents; nonetheless, mouse models of lung fibrosis provide clues as to the natural progression of lung injury and remodeling, but many have not been useful in predicting efficacy of therapeutics in clinical IPF. We provide a detailed methodologic description of various iterations of humanized mouse models, initiated by the i.v. injection of cells from IPF lung biopsy or explants specimens into severe combined immunodeficiency (SCID)/beige or nonobese diabetic SCID γ mice. Unlike cells from normal lung samples, IPF cells promote persistent, nonresolving lung remodeling in SCID mice. Finally, we provide examples and discuss potential advantages and pitfalls of human-specific targeting approaches in a humanized SCID model of pulmonary fibrosis.
Collapse
Affiliation(s)
- David M Habiel
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Milena S Espindola
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ana L Coelho
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Cory M Hogaboam
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
37
|
Abstract
The objective of this study was to determine whether urine ubiquitin levels are elevated after burns and to assess whether urine ubiquitin could be useful as a noninvasive biomarker for burn patients. Forty burn patients (%TBSA: 20 ± 22; modified Baux scores: 73 ± 26) were included (control: 11 volunteers). Urine was collected in 2-hour intervals for 72 hours, followed by 12-hour intervals until discharge from the intensive care unit. Ubiquitin concentrations were analyzed by enzyme linked immunosorbent assay and Western blot. Total protein was determined with a Bradford assay. Patient characteristics and clinical parameters were documented. Urine ubiquitin concentrations, renal ubiquitin excretion, and excretion rates were correlated with patient characteristics and outcomes. Initial urine ubiquitin concentrations were 362 ± 575 ng/ml in patients and 14 ± 18 ng/ml in volunteers (P < .01). Renal ubiquitin excretion on day 1 was 292.6 ± 510.8 μg/24 hr and 21 ± 27 μg/24 hr in volunteers (P < .01). Initial ubiquitin concentrations correlated with modified Baux scores (r = .46; P = .02). Ubiquitin levels peaked at day 6 postburn, whereas total protein concentrations and serum creatinine levels remained within the normal range. Total renal ubiquitin excretion and excretion rates were higher in patients with %TBSA ≥20 than with %TBSA <20, in patients who developed sepsis/multiple organ failure than in patients without these complications and in nonsurvivors vs survivors. These data suggest that ubiquitin urine levels are significantly increased after burns. Renal ubiquitin excretion and/or excretion rates are associated with %TBSA, sepsis/multiple organ failure, and mortality. Although these findings may explain previous correlations between systemic ubiquitin levels and outcomes after burns, the large variability of ubiquitin urine levels suggests that urine ubiquitin will not be useful as a noninvasive disease biomarker.
Collapse
|
38
|
Griffiths K, Habiel DM, Jaffar J, Binder U, Darby WG, Hosking CG, Skerra A, Westall GP, Hogaboam CM, Foley M. Anti-fibrotic Effects of CXCR4-Targeting i-body AD-114 in Preclinical Models of Pulmonary Fibrosis. Sci Rep 2018; 8:3212. [PMID: 29453386 PMCID: PMC5816662 DOI: 10.1038/s41598-018-20811-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/24/2018] [Indexed: 01/19/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibrotic lung disease that is prevalent in individuals >50 years of age, with a median survival of 3–5 years and limited therapeutic options. The disease is characterized by collagen deposition and remodeling of the lung parenchyma in a process that is thought to be driven by collagen-expressing immune and structural cells. The G-protein coupled C-X-C chemokine receptor 4, CXCR4, is a candidate therapeutic target for IPF owing to its role in the recruitment of CXCR4+ fibrocytes from the bone marrow to fibrotic lung tissue and its increased expression levels by structural cells in fibrotic lung tissue. We have engineered a novel fully human single domain antibody “i-body” called AD-114 that binds with high affinity to human CXCR4. We demonstrate here that AD-114 inhibits invasive wound healing and collagen 1 secretion by human IPF fibroblasts but not non-diseased control lung fibroblasts. Furthermore, in a murine bleomycin model of pulmonary fibrosis, AD-114 reduced the accumulation of fibrocytes (CXCR4+/Col1+/CD45+) in fibrotic murine lungs and ameliorated the degree of lung injury. Collectively, these studies demonstrate that AD-114 holds promise as a new biological therapeutic for the treatment of IPF.
Collapse
Affiliation(s)
- K Griffiths
- AdAlta Limited, La Trobe University, 15/2 Park Drive, Bundoora, 3083, Australia.,The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, 3086, Australia
| | - D M Habiel
- Cedars-Sinai, Medical Centre, Los Angeles, CA, 90048, USA
| | - J Jaffar
- Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, 3000, Australia
| | - U Binder
- XL-protein GmbH, Lise-Meitner-Str. 30, 85354, Freising, Germany
| | - W G Darby
- AdAlta Limited, La Trobe University, 15/2 Park Drive, Bundoora, 3083, Australia.,The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, 3086, Australia
| | - C G Hosking
- AdAlta Limited, La Trobe University, 15/2 Park Drive, Bundoora, 3083, Australia.,The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, 3086, Australia
| | - A Skerra
- XL-protein GmbH, Lise-Meitner-Str. 30, 85354, Freising, Germany
| | - G P Westall
- Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, 3000, Australia
| | - C M Hogaboam
- Cedars-Sinai, Medical Centre, Los Angeles, CA, 90048, USA
| | - M Foley
- AdAlta Limited, La Trobe University, 15/2 Park Drive, Bundoora, 3083, Australia. .,The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, 3086, Australia.
| |
Collapse
|
39
|
Lee Y, Blount KL, Dai F, Thompson S, Scher JK, Bitterman S, Droher M, Herzog EL, Moeckel G, Karihaloo A, Dahl NK. Semaphorin 7A in circulating regulatory T cells is increased in autosomal-dominant polycystic kidney disease and decreases with tolvaptan treatment. Clin Exp Nephrol 2018; 22:906-916. [PMID: 29453607 DOI: 10.1007/s10157-018-1542-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 02/07/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Semaphorin 7A (SEMA7A) is an immunomodulating protein implicated in lung and liver fibrosis. In autosomal-dominant polycystic kidney disease (ADPKD), the progressive expansion of renal cysts, inflammation, and subsequent renal fibrosis leads to end-stage renal disease (ESRD). SEMA7A may play a role in renal fibrosis and in ADPKD. METHODS We evaluated Sema7a in a mouse model of renal fibrosis and determined the expression of SEMA7A in human ADPKD kidney. We analyzed SEMA7A expression on peripheral blood mononuclear cells (PBMCs), including CD45+ (leukocyte), CD14+(monocyte), CD4+ (T lymphocytes) and CD4+Foxp3+CD25+ [regulatory T lymphocytes (Tregs)] from 90 ADPKD patients (11 tolvaptan treated and 79 tolvaptan naïve), and 21 healthy volunteers, using a Fluorescence-Activated Cell Sorting (FACS). RESULTS Sema7a is required for renal fibrosis. SEMA7A shows robust expression in ADPKD kidneys, localizing to cysts derived from distal tubules. SEMA7A is higher in circulating monocytes, but unchanged in CD4+ lymphocytes in ADPKD patients. The SEMA7A increase was detected early (stage 1 CKD) and seemed more prominent in patients with smaller kidneys (p = 0.09). Compared to tolvaptan-naïve ADPKD patients, those treated with tolvaptan showed reduced SEMA7A expression on monocytes, T lymphocytes, and Tregs, although the number of PBMCs was unchanged. After 1 month of tolvaptan treatment, SEMA7A expression on Tregs decreased. CONCLUSIONS SEMA7A shows potential as both a therapeutic target in mammalian kidney fibrosis and as a marker of inflammation in ADPKD patients. SEMA7A expression was lower after tolvaptan treatment, which may reflect drug efficacy.
Collapse
Affiliation(s)
- Yashang Lee
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | | | - Feng Dai
- Department of Biostatics, Yale University School of Public Health, 300 George Street, New Haven, CT, 06511, USA
| | - Siobhan Thompson
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | | | - Sherrie Bitterman
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Madeline Droher
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Erica L Herzog
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Gilbert Moeckel
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Anil Karihaloo
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Neera K Dahl
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
40
|
Jackson EK, Zhang Y, Gillespie DD, Zhu X, Cheng D, Jackson TC. SDF-1α (Stromal Cell-Derived Factor 1α) Induces Cardiac Fibroblasts, Renal Microvascular Smooth Muscle Cells, and Glomerular Mesangial Cells to Proliferate, Cause Hypertrophy, and Produce Collagen. J Am Heart Assoc 2017; 6:JAHA.117.007253. [PMID: 29114002 PMCID: PMC5721794 DOI: 10.1161/jaha.117.007253] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Activated cardiac fibroblasts (CFs), preglomerular vascular smooth muscle cells (PGVSMCs), and glomerular mesangial cells (GMCs) proliferate, cause hypertrophy, and produce collagen; in this way, activated CFs contribute to cardiac fibrosis, and activated PGVSMCs and GMCs promote renal fibrosis. In heart and kidney diseases, SDF-1α (stromal cell-derived factor 1α; endogenous CXCR4 [C-X-C motif chemokine receptor 4] receptor agonist) levels are often elevated; therefore, it is important to know whether and how the SDF-1α/CXCR4 axis activates CFs, PGVSMCs, or GMCs. METHODS AND RESULTS Here we investigated whether SDF-1α activates CFs, PGVSMCs, and GMCs to proliferate, hypertrophy, or produce collagen. DPP4 (dipeptidyl peptidase 4) inactivates SDF-1α and previous experiments show that growth-promoting peptides have greater effects in cells from genetically-hypertensive animals. Therefore, we performed experiments in the absence and presence of sitagliptin (DPP4 inhibitor) and in cells from normotensive Wistar-Kyoto rats and spontaneously hypertensive rats. Our studies show (1) that spontaneously hypertensive and Wistar-Kyoto rat CFs, PGVSMCs, and GMCs express CXCR4 receptors and DPP4 activity; (2) that chronic treatment with physiologically relevant concentrations of SDF-1α causes concentration-dependent increases in the proliferation (cell number) and hypertrophy (3H-leucine incorporation) of and collagen production (3H-proline incorporation) by CFs, PGVSMCs, and GMCs; (3) that sitagliptin augments these effects of SDF-1α; (4) that interactions between SDF-1α and sitagliptin are greater in spontaneously hypertensive rat cells; (5) that CXCR4 antagonism (AMD3100) blocks all effects of SDF-1α; and (6) that SDF-1α/CXCR4 signal transduction likely involves the RACK1 (receptor for activated C kinase 1)/Gβγ/PLC (phospholipase C)/PKC (protein kinase C) signaling complex. CONCLUSIONS The SDF-1α/CXCR4 axis drives proliferation and hypertrophy of and collagen production by CFs, PGVSMCs, and GMCs, particularly in cells from genetically hypertensive animals and when DPP4 is inhibited.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Yumeng Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Delbert D Gillespie
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Xiao Zhu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Dongmei Cheng
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Travis C Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
41
|
Abstract
Acute kidney injury (AKI) is a growing global health concern, yet no treatment is currently available to prevent it or to promote kidney repair after injury. Animal models demonstrate that the macrophage is a major contributor to the inflammatory response to AKI. Emerging data from human biopsies also corroborate the presence of macrophages in AKI and their persistence in progressive chronic kidney disease. Macrophages are phagocytic innate immune cells that are important mediators of tissue homeostasis and host defense. In response to tissue injury, macrophages become activated based on specific signals from the damaged microenvironment. The activation and functional state of the macrophage depends on the stage of tissue injury and repair, reflecting a dynamic and diverse spectrum of macrophage phenotypes. In this review, we highlight our current understanding of the mechanisms by which macrophages contribute to injury and repair after AKI.
Collapse
Affiliation(s)
- Sarah C Huen
- Section of Nephrology, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520;
| | - Lloyd G Cantley
- Section of Nephrology, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520;
| |
Collapse
|
42
|
CCR2 contributes to the recruitment of monocytes and leads to kidney inflammation and fibrosis development. Inflammopharmacology 2017; 26:403-411. [PMID: 28168553 DOI: 10.1007/s10787-017-0317-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 01/23/2017] [Indexed: 12/24/2022]
Abstract
Chemokines are a large family of proteins that, once associated to its receptor on leukocytes, stimulate their movement and migration from blood to tissues. Once in the tissue, immune cells trigger inflammation that, when uncontrolled, leads to fibrosis development. Among the immune cells, macrophages take a special role in fibrosis formation, since macrophage depletion reflects less collagen deposition. The majority of tissue macrophages is derived from monocytes, especially monocytes expressing the chemokine receptor CCR2. Here, we investigated the role of infiltrating CCR2+ cells in the development of fibrosis, and specifically, the dynamic of infiltration of these cells into kidneys under chronic obstructive lesion. Using liposome-encapsulated clodronate, we observed that macrophage depletion culminated in less collagen deposition and reduced chemokines milieu that were released in the damaged kidney after obstructive nephropathy. We also obstructed the kidneys of CCL3-/-, CCR2-/-, CCR4-/-, CCR5-/-, and C57BL/6 mice and we found that among all animals, CCR2-/- mice demonstrated the more robust protection, reflected by less inflammatory and Th17-related cytokines and less collagen formation. Next we evaluated the dynamic of CCR2+/rfp cell infiltration and we observed that they adhere onto the vessels at early stages of disease, culminating in increased recruitment of CCR2+/rfp cells at later stages. On the other hand, CCR2rfp/rfp animals exhibited less fibrosis formation and reduced numbers of recruited cells at later stages. We have experimentally demonstrated that inflammatory CCR2+ cells that reach the injured kidney at initial stages after tissue damage are responsible for the fibrotic pattern observed at later time points in the context of UUO.
Collapse
|
43
|
Expression of chemokine receptors on peripheral blood T cells in children with chronic kidney disease. Mediators Inflamm 2015; 2015:536894. [PMID: 25866451 PMCID: PMC4381676 DOI: 10.1155/2015/536894] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 02/18/2015] [Accepted: 02/27/2015] [Indexed: 12/31/2022] Open
Abstract
Chemokine receptors play a role in leukocyte recruitment, activation, and maintaining effector functions and regulate adaptive immune response and angiogenesis. The study aimed at flow cytometric analysis of T cell subsets with selected surface chemokine receptors (CCR4, CCR5, CCR7, CXCR3, and CXCR4) or receptor combination in peripheral blood of children with chronic kidney disease (CKD) on hemodialysis (HD). The percentage of T lymphocytes with CD8 and combined CD28,CCR7 expression was higher in HD children. The percentage of T lymphocytes expressing CCR7, CD28,CCR7, and CXCR4,CD8 was increased in children on conservative treatment. Total number (tn) of CXCR4+ cells was reduced in children on hemodialysis. The tn of T CXCR3+ cells was lower in children on conservative treatment. During HD the percentage of T CD4+ cells was higher and of T CXCR3+ lymphocytes was lower after HD session as compared to 15 min of session duration. During HD tn of T cells with expression of CCR4, CCR5, CCR7, CXCR3, and CXCR4 was constant. The alteration of chemokine receptors expression in children with CKD occurs early in the development. Diminished expression of CXCR3, CXCR4 on T cells in patients with CKD on HD might result in impaired inflammatory response. Increased CCR7+ T cell percentage could be responsible for the alteration of migration of cells into secondary lymphatic organs.
Collapse
|