1
|
Dev S, Dong Y, Hamilton JP. Hepatic microtubule destabilization facilitates liver fibrosis in the mouse model of Wilson disease. J Mol Med (Berl) 2025; 103:531-545. [PMID: 40140071 PMCID: PMC12078373 DOI: 10.1007/s00109-025-02535-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
Wilson disease (WD) is a potentially fatal metabolic disorder caused by the inactivation of the copper (Cu) transporter ATP7B, resulting in systemic Cu overload and fibroinflammatory liver disease. The molecular mechanism and effects of elevated Cu on cytoskeletal dynamics in liver fibrogenesis are not clear. Here, we tested the regulation of hepatic cytoskeleton and fibrogenesis with respect to Cu overload in WD. Atp7b-/- (knockout) mice with established liver disease, hepatocyte-specific Atp7b△Hep knockout mice without fibroinflammatory disease, and the age-and sex-matched controls were compared using Western blotting, real-time quantitative reverse transcription PCR (qRT-PCR), immunohistochemical (IHC) staining and transcriptomics (RNA-sequencing) analysis. In Atp7b-/- mice with developed liver disease, there is a significant increase in cytoskeletal protein expression with a reduction in α-tubulin acetylation. In these mice before the onset of liver pathology, no significant changes in cytoskeletal nor hepatic stellate cell activation are observed. As hepatic copper levels rise, an increase in cytoskeletal proteins with a decrease in acetylated-α-tubulin/α-tubulin ratio occurs. RNA-sequencing, qRT-PCR, and immunostaining confirm that the tubulin is upregulated at the transcriptional level and hepatocytes are the primary source of early tubulin increases before fibrosis. An increase in α-tubulin with a decrease in α-tubulin acetylation via Hdac6 and Sirt2 induction facilitates fibrosis as reflected by concomitant increases in desmin and α-SMA immunostaining in Atp7b-/- mice at 20 weeks. Moreover, strongly positive correlations between α-tubulin and α-tubulin deacetylase with the expression of liver fibrosis markers are observed in animal and human WD. Hepatocyte-specific Atp7b△Hep mice lack significant changes in tubulin as well as fibrosis despite hepatic steatosis. This study provides evidence that microtubule destabilization causes cytoskeletal rearrangement and facilitates hepatic stellate cell (HSC) activation and fibrosis in the murine model of WD. KEY MESSAGES: Hepatic cytoskeleton system is induced in Wilson disease. Hepatic microtubules acetylation is dysregulated in murine Wilson disease. Microtubules destabilization is positively associated with liver fibrosis in Wilson disease. Microtubules destabilization concomitant with fibrogenesis exacerbates WD progression.
Collapse
Affiliation(s)
- Som Dev
- Department of Biochemistry, All India Institute of Medical Sciences, Kalyani, West Bengal, 741245, India.
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| | - Yixuan Dong
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - James P Hamilton
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Division of Gastroenterology and Hepatology, Johns Hopkins University, School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
| |
Collapse
|
2
|
Huang H, Han Y, Zhang Y, Zeng J, He X, Cheng J, Wang S, Xiong Y, Yin H, Yuan Q, Huang L, Xie Y, Meng J, Tao L, Peng Z. Deletion of Pyruvate Carboxylase in Tubular Epithelial Cell Promotes Renal Fibrosis by Regulating SQOR/cGAS/STING-Mediated Glycolysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408753. [PMID: 39836535 PMCID: PMC11967762 DOI: 10.1002/advs.202408753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 12/31/2024] [Indexed: 01/23/2025]
Abstract
Renal fibrosis is a common pathway involved in the progression of various chronic kidney diseases to end-stage renal disease. Recent studies show that mitochondrial injury of renal tubular epithelial cells (RTECs) is a crucial pathological foundation for renal fibrosis. However, the underlying regulatory mechanisms remain unclear. Pyruvate carboxylase (PC) is a catalytic enzyme located within the mitochondria that is intricately linked with mitochondrial damage and metabolism. In the present study, the downregulation of PC in various fibrotic animal and human kidney samples is demonstrated. Renal proximal tubule-specific Pcx gene knockout mice (PcxcKO) has significant interstitial fibrosis compared to control mice, with heightened expression of extracellular matrix molecules. This is further demonstrated in a stable PC knock-out RTEC line. Mechanistically, PC deficiency reduces its interaction with sulfide:quinone oxidoreductase (SQOR), increasing the ubiquitination and degradation of SQOR. This leads to mitochondrial morphological and functional disruption, increased mtDNA release, activation of the cGAS-STING pathway, and elevated glycolysis levels, and ultimately, promotes renal fibrosis. This study investigates the molecular mechanisms through which PC deficiency induces mitochondrial injury and metabolic reprogramming in RTECs. This study provides a novel theoretical foundation and potential therapeutic targets for the pathogenesis and treatment of renal fibrosis.
Collapse
Affiliation(s)
- Hao Huang
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Department of Cell biologySchool of Life SciencesCentral South UniversityChangsha410013China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
| | - Yuanyuan Han
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Yan Zhang
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Jianhua Zeng
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Xin He
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Jiawei Cheng
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Songkai Wang
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Yiwei Xiong
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Hongling Yin
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- Department of Pathology, Xiangya HospitalCentral South UniversityChangsha410008China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Ling Huang
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Yanyun Xie
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Jie Meng
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- Department of Pulmonary and Critical Care Medicine, Third Xiangya HospitalCentral South UniversityChangsha410013China
| | - Lijian Tao
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya HospitalCentral South UniversityChangsha410008China
- Hunan Key Laboratory of Organ FibrosisCentral South UniversityChangsha410013China
- FuRong LaboratoryChangsha410008China
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangsha410008China
- National Medical Metabolomics International Collaborative Research CenterCentral South UniversityChangsha410008China
| |
Collapse
|
3
|
Fang X, Zhong Y, Zheng R, Wu Q, Liu Y, Zhang D, Wang Y, Ding W, Wang K, Zhong F, Lin K, Yao X, Hu Q, Li X, Xu G, Liu N, Nie J, Li D, Geng H, Guan Y. PPDPF preserves integrity of proximal tubule by modulating NMNAT activity in chronic kidney diseases. SCIENCE ADVANCES 2025; 11:eadr8648. [PMID: 40106551 PMCID: PMC11922016 DOI: 10.1126/sciadv.adr8648] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/24/2024] [Indexed: 03/22/2025]
Abstract
Genome-wide association studies (GWAS) have identified loci associated with kidney diseases, but the causal variants, genes, and pathways involved remain elusive. Here, we identified a kidney disease gene called pancreatic progenitor cell differentiation and proliferation factor (PPDPF) through integrating GWAS on kidney function and multiomic analysis. PPDPF was predominantly expressed in healthy proximal tubules of human and mouse kidneys via single-cell analysis. Further investigations revealed that PPDPF functioned as a thiol-disulfide oxidoreductase to maintain cellular NAD+ levels. Deficiency in PPDPF disrupted NAD+ and mitochondrial homeostasis by impairing the activities of nicotinamide mononucleotide adenylyl transferases (NMNATs), thereby compromising the function of proximal tubules during injuries. Consequently, knockout of PPDPF notably accelerated the progression of chronic kidney disease (CKD) in mouse models induced by aging, chemical exposure, and obstruction. These findings strongly support targeting PPDPF as a potential therapy for kidney fibrosis, offering possibilities for future CKD interventions.
Collapse
Affiliation(s)
- Xiaoliang Fang
- Department of Urology, Children’s Hospital of Fudan University, Shanghai, 201102, China
| | - Yi Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Rui Zheng
- Department of Urology, Children’s Hospital of Fudan University, Shanghai, 201102, China
| | - Qihui Wu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Yu Liu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Dexin Zhang
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuwei Wang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Wubing Ding
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kaiyuan Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fengbo Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kai Lin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiaohui Yao
- Qingdao Innovation and Development Center, Harbin Engineering University, Qingdao, Shandong, 266000, China
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, Heilongjiang, 150001, China
| | - Qingxun Hu
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiaofei Li
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, 17164, Sweden
| | - Guofeng Xu
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jing Nie
- Biobank of Peking University First Hospital, Peking University First Hospital, Peking University, Beijing, 100034, China
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Hongquan Geng
- Department of Urology, Children’s Hospital of Fudan University, Shanghai, 201102, China
| | - Yuting Guan
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China
| |
Collapse
|
4
|
Zhang S, Liu K, Liu Y, Hu X, Gu X. The role and application of bioinformatics techniques and tools in drug discovery. Front Pharmacol 2025; 16:1547131. [PMID: 40017606 PMCID: PMC11865229 DOI: 10.3389/fphar.2025.1547131] [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: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
The process of drug discovery and development is both lengthy and intricate, demanding a substantial investment of time and financial resources. Bioinformatics techniques and tools can not only accelerate the identification of drug targets and the screening and refinement of drug candidates, but also facilitate the characterization of side effects and the prediction of drug resistance. High-throughput data from genomics, transcriptomics, proteomics, and metabolomics make significant contributions to mechanics-based drug discovery and drug reuse. This paper summarizes bioinformatics technologies and tools in drug research and development and their roles and applications in drug research and development, aiming to provide references for the development of new drugs and the realization of precision medicine.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Kaijie Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yafeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Xinjun Hu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Medical Key Laboratory of Gastrointestinal Microecology and Hepatology, Luoyang, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
5
|
Sun L, Wang C, Zhou Z, Li Q. An integrated proteomic and phosphoproteomic landscape of chronic kidney disease. J Proteomics 2025; 311:105355. [PMID: 39547397 DOI: 10.1016/j.jprot.2024.105355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/27/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
The prevalence of chronic kidney disease (CKD) is gradually rising worldwide. Patients often remain asymptomatic for an extended period, leaving them unaware of their condition, which can lead to progressing to end-stage renal disease and cause significant economic burden. Improved understanding of CKD pathogenesis can enhance early detection and facilitate advances in drug development. Here, we performed proteomic and phosphoproteomic analyses of the mouse unilateral ureteral obstruction model to explore the molecular mechanisms of chronic kidney injury. 474 significantly differentially expressed proteins and 96 significantly differentially expressed phosphoproteins were screened, respectively. Chronic kidney injury involves complex metabolic pathways such as citrate cycle and hematopoietic system in proteome, and mitochondrial oxidative phosphorylation suppression is a notable alteration. The phosphoproteomic analysis revealed a significant upregulation in epithelial mesenchymal transition and P53 pathways, with a corresponding increase in the phosphorylation of Jun at serine 73. Utilizing HK2 cells, we observed that the reduction oxidative phosphorylation was consistently associated with an augmentation in oxidative stress, which subsequently activated Jun and induced apoptosis. Proteins that act as hubs in these pathways may be candidate targets for CKD intervention. These findings contribute significantly to the current understanding of CKD and provide valuable insights for future studies. SIGNIFICANCE: Chronic kidney disease (CKD) incidence rising annually with varied etiologies, kidney often irreversibly fibrotic, the treatment options are limited and often ineffective due to deficient understanding of renal fibrosis mechanisms. Despite the extensive efforts and numerous omics studies conducted on renal fibrosis, to date, no study has been undertaken to investigate the role of phosphorylated proteins in UUO models. Previously, we performed a comprehensive transcriptome and proteome analysis based on the CKD model, but the potential alterations in the phosphoproteome were not addressed. Here, an integrated proteomic and phosphoproteomic landscape of CKD was completed, which was the the first phosphoproteomic profiles of UUO model. Phosphoproteomic profile suggests that the epithelial mesenchymal transition and P53 pathways is significantly activated in mouse models of kidney injury, and the core protein Jun played a key role in CKD. And a preliminary correlation between P-Jun and oxidative phosphorylation was found base on HK2 cells. Our work contributes to a deeper understanding of the disease characteristics and molecular mechanisms of CKD. Identifying potential CKD targets from proteome and phosphoproteome may provide valuable insights for early diagnosis and treatment of CKD.
Collapse
Affiliation(s)
- Linxiao Sun
- Department of Laboratory, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang 325000, China
| | - Cheng Wang
- Department of Laboratory, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, China
| | - Zhongjing Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Qiangqiang Li
- Department of General Surgery, the People's Hospital of Yuhuan, Taizhou, Zhejiang 317600, China.
| |
Collapse
|
6
|
Stepanova G, Manzéger A, Mózes MM, Kökény G. Renal Epithelial Complement C3 Expression Affects Kidney Fibrosis Progression. Int J Mol Sci 2024; 25:12551. [PMID: 39684261 DOI: 10.3390/ijms252312551] [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: 09/24/2024] [Revised: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Kidney fibrosis is a hallmark of chronic kidney diseases. Evidence shows that genetic variability and complement component 3 (C3) might influence tubulointerstitial fibrosis. Still, the role of renal C3 production in the epithelial-to-mesenchymal transition (EMT) and genetically determined fibrosis progression remains undiscovered. The kidneys of fibrosis-resistant C57Bl/6J (B6) and fibrosis-prone CBA/J (CBA) and BALB/cJ (BalbC) mice (n = 4-8/group) were subjected to unilateral ureteral obstruction (UUO) and analyzed after 1, 3, and 7 days, along with human focal glomerular sclerotic (FSGS) and healthy kidneys. Mouse primary tubular epithelial cells (PTECs) were investigated after 24 h of treatment with transforming growth factor β (TGFβ) or complement anaphylatoxin 3a (C3a) agonist (n = 4/group). UUO resulted in delayed kidney injury in fibrosis-resistant B6 mice, but very early renal C3 messenger RNA (mRNA) induction in fibrosis-prone CBA and BalbC mice, along with collagen I (Col1a1) and collagen III (Col3a1). CBA depicted the fastest fibrosis progression with the highest C3, lipocalin-2 (Lcn2), Tgfb1, and chemokine (C-C motif) ligand 2 (Ccl2) expression. Human FSGS kidneys depicted C3 mRNA over-expression and strong tubular C3 immunostaining. In PTECs, C3a agonist treatment induced pro-fibrotic early growth response protein 1 (EGR1) expression and the EMT, independent of TGFβ signaling. We conclude that de novo renal tubular C3 synthesis is associated with the genetically determined kidney fibrosis progression rate in mice and the pathogenesis of FSGS in humans. This tubular C3 overproduction can, through local pro-fibrotic effects, influence the progression of chronic kidney disease.
Collapse
Affiliation(s)
- Ganna Stepanova
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Anna Manzéger
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
- International Nephrology Research and Training Center, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Miklós M Mózes
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
- International Nephrology Research and Training Center, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Gábor Kökény
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
- International Nephrology Research and Training Center, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| |
Collapse
|
7
|
Okamoto Y, Kitakaze K, Takenouchi Y, Matsui R, Koga D, Miyashima R, Ishimaru H, Tsuboi K. GPR176 promotes fibroblast-to-myofibroblast transition in organ fibrosis progression. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119798. [PMID: 39047914 DOI: 10.1016/j.bbamcr.2024.119798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/20/2024] [Accepted: 07/14/2024] [Indexed: 07/27/2024]
Abstract
Fibrosis is characterized by excessive deposition of extracellular matrix proteins, particularly collagen, caused by myofibroblasts in response to chronic inflammation. Although G protein-coupled receptors (GPCRs) are among the targets of current antifibrotic drugs, no drug has yet been approved to stop fibrosis progression. Herein, we aimed to identify GPCRs with profibrotic effects. In gene expression analysis of mouse lungs with induced fibrosis, eight GPCRs were identified, showing a >2-fold increase in mRNA expression after fibrosis induction. Among them, we focused on Gpr176 owing to its significant correlation with a myofibroblast marker α-smooth muscle actin (αSMA), the profibrotic factor transforming growth factor β1 (TGFβ1), and collagen in a human lung gene expression database. Similar to the lung fibrosis model, increased Gpr176 expression was also observed in other organs affected by fibrosis, including the kidney, liver, and heart, suggesting its role in fibrosis across various organs. Furthermore, fibroblasts abundantly expressed Gpr176 compared to alveolar epithelial cells, endothelial cells, and macrophages in the fibrotic lung. GPR176 expression was unaffected by TGFβ1 stimulation in rat renal fibroblast NRK-49 cells, whereas knockdown of Gpr176 by siRNA reduced TGFβ1-induced expression of αSMA, fibronectin, and collagen as well as Smad2 phosphorylation. This suggested that Gpr176 regulates fibroblast activation. Consequently, Gpr176 acts in a profibrotic manner, and inhibiting its activity could potentially prevent myofibroblast differentiation and improve fibrosis. Developing a GPR176 inverse agonist or allosteric modulator is a promising therapeutic approach for fibrosis.
Collapse
Affiliation(s)
- Yasuo Okamoto
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan.
| | - Keisuke Kitakaze
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Yasuhiro Takenouchi
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Rena Matsui
- Department of Medical Technology, Kawasaki University of Medical Welfare, Kurashiki, Okayama 701-0192, Japan
| | - Daisuke Koga
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Ryo Miyashima
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Hironobu Ishimaru
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| | - Kazuhito Tsuboi
- Department of Pharmacology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan
| |
Collapse
|
8
|
Hu Y, Huang H, Xiang R. GCGR: novel potential therapeutic target for chronic kidney disease. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1542-1544. [PMID: 38679668 DOI: 10.1007/s11427-024-2576-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/22/2024] [Indexed: 05/01/2024]
Affiliation(s)
- Yan Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Huang
- Department of Nephrology, Xiangya Hospital Central South University, Changsha, 410008, China.
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, 410013, China.
| | - Rong Xiang
- Department of Nephrology, Xiangya Hospital Central South University, Changsha, 410008, China.
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China.
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, 410013, China.
| |
Collapse
|
9
|
Cheng DH, Jiang TG, Zeng WB, Li TM, Jing YD, Li ZQ, Guo YH, Zhang Y. Identification and coregulation pattern analysis of long noncoding RNAs in the mouse brain after Angiostrongylus cantonensis infection. Parasit Vectors 2024; 17:205. [PMID: 38715092 PMCID: PMC11077716 DOI: 10.1186/s13071-024-06278-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Angiostrongyliasis is a highly dangerous infectious disease. Angiostrongylus cantonensis larvae migrate to the mouse brain and cause symptoms, such as brain swelling and bleeding. Noncoding RNAs (ncRNAs) are novel targets for the control of parasitic infections. However, the role of these molecules in A. cantonensis infection has not been fully clarified. METHODS In total, 32 BALB/c mice were randomly divided into four groups, and the infection groups were inoculated with 40 A. cantonensis larvae by gavage. Hematoxylin and eosin (H&E) staining and RNA library construction were performed on brain tissues from infected mice. Differential expression of long noncoding RNAs (lncRNAs) and mRNAs in brain tissues was identified by high-throughput sequencing. The pathways and functions of the differentially expressed lncRNAs were determined by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. The functions of the differentially expressed lncRNAs were further characterized by lncRNA‒microRNA (miRNA) target interactions. The potential host lncRNAs involved in larval infection of the brain were validated by quantitative real-time polymerase chain reaction (qRT‒PCR). RESULTS The pathological results showed that the degree of brain tissue damage increased with the duration of infection. The transcriptome results showed that 859 lncRNAs and 1895 mRNAs were differentially expressed compared with those in the control group, and several lncRNAs were highly expressed in the middle-late stages of mouse infection. GO and KEGG pathway analyses revealed that the differentially expressed target genes were enriched mainly in immune system processes and inflammatory response, among others, and several potential regulatory networks were constructed. CONCLUSIONS This study revealed the expression profiles of lncRNAs in the brains of mice after infection with A. cantonensis. The lncRNAs H19, F630028O10Rik, Lockd, AI662270, AU020206, and Mexis were shown to play important roles in the infection of mice with A. cantonensis infection.
Collapse
Affiliation(s)
- Dong-Hui Cheng
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China
| | - Tian-Ge Jiang
- School of Global Health, National Center for Tropical Disease Research, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wen-Bo Zeng
- School of Life Sciences, Fudan University, Shanghai, People's Republic of China
| | - Tian-Mei Li
- Dali Prefectural Institute of Research and Control On Schistosomiasis, Yunnan, People's Republic of China
| | - Yi-Dan Jing
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China
| | - Zhong-Qiu Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China
| | - Yun-Hai Guo
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China
| | - Yi Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (National Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases; WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, People's Republic of China.
- School of Global Health, National Center for Tropical Disease Research, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| |
Collapse
|
10
|
Wang C, Pan Z, Sun L, Li Q. Integrative transcriptomic and proteomic profile revealed inhibition of oxidative phosphorylation and peroxisomes during renal interstitial fibrosis. J Proteomics 2024; 298:105144. [PMID: 38431085 DOI: 10.1016/j.jprot.2024.105144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
Effective therapies of chronic kidney disease (CKD) are lacking due to the unclear molecular pathogenesis. Previous single omics-studies have described potential molecular regulation mechanism of CKD only at the level of transcription or translation. Therefore, this study generated an integrated transcriptomic and proteomic profile to provide deep insights into the continuous transcription-translation process during CKD. The comprehensive datasets identified 14,948 transcripts and 6423 proteins, 233 up-regulated and 364 down-regulated common differentially expressed genes of transcriptome and proteome were selected to further combined bioinformatics analysis. The obtained results revealed reactive oxygen species (ROS) metabolism and antioxidant system due to imbalance of mitochondria and peroxisomes were significantly repressed in CKD. Overall, this study presents a valuable multi-omics analysis that sheds light on the molecular mechanisms underlying CKD. SIGNIFICANCE: Chronic kidney disease (CKD) is a progressive and irreversible condition that results in abnormal kidney function and structure, and is ranked 18th among the leading causes of death globally, leading to a significant societal burden. Hence, there is an urgent need for research to detect new, sensitive, and specific biomarkers. Omics-based studies offer great potential to identify underlying disease mechanisms, aid in clinical diagnosis, and develop novel treatment strategies for CKD. Previous studies have mainly focused on the regulation of gene expression or protein synthesis in CKD, thereby compelling us to conduct a meticulous analysis of transcriptomic and proteomic data from the UUO mouse model. Here, we have performed a unified analysis of CKD model by integrating transcriptomes and protein suites for the first time. Our study contributes to a deeper understanding of the pathogenesis of CKD and provides a basis for subsequent disease management and drug development.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Laboratory, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, PR China
| | - Zhuo Pan
- Department of General Surgery, First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, PR China
| | - Linxiao Sun
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang 325000, PR China
| | - Qiangqiang Li
- Department of General Surgery, the People's Hospital of Yuhuan, Taizhou 317600, Zhejiang, PR China.
| |
Collapse
|
11
|
Landsberger T, Amit I, Alon U. Geroprotective interventions converge on gene expression programs of reduced inflammation and restored fatty acid metabolism. GeroScience 2024; 46:1627-1639. [PMID: 37698783 PMCID: PMC10828297 DOI: 10.1007/s11357-023-00915-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/15/2023] [Indexed: 09/13/2023] Open
Abstract
Understanding the mechanisms of geroprotective interventions is central to aging research. We compare four prominent interventions: senolysis, caloric restriction, in vivo partial reprogramming, and heterochronic parabiosis. Using published mice transcriptomic data, we juxtapose these interventions against normal aging. We find a gene expression program common to all four interventions, in which inflammation is reduced and several metabolic processes, especially fatty acid metabolism, are increased. Normal aging exhibits the inverse of this signature across multiple organs and tissues. A similar inverse signature arises in three chronic inflammation disease models in a non-aging context, suggesting that the shift in metabolism occurs downstream of inflammation. Chronic inflammation is also shown to accelerate transcriptomic age. We conclude that a core mechanism of geroprotective interventions acts through the reduction of inflammation with downstream effects that restore fatty acid metabolism. This supports the notion of directly targeting genes associated with these pathways to mitigate age-related deterioration.
Collapse
Affiliation(s)
- Tomer Landsberger
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
12
|
Reiss AB, Jacob B, Zubair A, Srivastava A, Johnson M, De Leon J. Fibrosis in Chronic Kidney Disease: Pathophysiology and Therapeutic Targets. J Clin Med 2024; 13:1881. [PMID: 38610646 PMCID: PMC11012936 DOI: 10.3390/jcm13071881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Chronic kidney disease (CKD) is a slowly progressive condition characterized by decreased kidney function, tubular injury, oxidative stress, and inflammation. CKD is a leading global health burden that is asymptomatic in early stages but can ultimately cause kidney failure. Its etiology is complex and involves dysregulated signaling pathways that lead to fibrosis. Transforming growth factor (TGF)-β is a central mediator in promoting transdifferentiation of polarized renal tubular epithelial cells into mesenchymal cells, resulting in irreversible kidney injury. While current therapies are limited, the search for more effective diagnostic and treatment modalities is intensive. Although biopsy with histology is the most accurate method of diagnosis and staging, imaging techniques such as diffusion-weighted magnetic resonance imaging and shear wave elastography ultrasound are less invasive ways to stage fibrosis. Current therapies such as renin-angiotensin blockers, mineralocorticoid receptor antagonists, and sodium/glucose cotransporter 2 inhibitors aim to delay progression. Newer antifibrotic agents that suppress the downstream inflammatory mediators involved in the fibrotic process are in clinical trials, and potential therapeutic targets that interfere with TGF-β signaling are being explored. Small interfering RNAs and stem cell-based therapeutics are also being evaluated. Further research and clinical studies are necessary in order to avoid dialysis and kidney transplantation.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (B.J.); (A.Z.); (A.S.); (M.J.); (J.D.L.)
| | | | | | | | | | | |
Collapse
|
13
|
Wang MY, Zhang Z, Zhao S, Onodera T, Sun XN, Zhu Q, Li C, Li N, Chen S, Paredes M, Gautron L, Charron MJ, Marciano DK, Gordillo R, Drucker DJ, Scherer PE. Downregulation of the kidney glucagon receptor, essential for renal function and systemic homeostasis, contributes to chronic kidney disease. Cell Metab 2024; 36:575-597.e7. [PMID: 38237602 PMCID: PMC10932880 DOI: 10.1016/j.cmet.2023.12.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 09/10/2023] [Accepted: 12/19/2023] [Indexed: 02/12/2024]
Abstract
The glucagon receptor (GCGR) in the kidney is expressed in nephron tubules. In humans and animal models with chronic kidney disease, renal GCGR expression is reduced. However, the role of kidney GCGR in normal renal function and in disease development has not been addressed. Here, we examined its role by analyzing mice with constitutive or conditional kidney-specific loss of the Gcgr. Adult renal Gcgr knockout mice exhibit metabolic dysregulation and a functional impairment of the kidneys. These mice exhibit hyperaminoacidemia associated with reduced kidney glucose output, oxidative stress, enhanced inflammasome activity, and excess lipid accumulation in the kidney. Upon a lipid challenge, they display maladaptive responses with acute hypertriglyceridemia and chronic proinflammatory and profibrotic activation. In aged mice, kidney Gcgr ablation elicits widespread renal deposition of collagen and fibronectin, indicative of fibrosis. Taken together, our findings demonstrate an essential role of the renal GCGR in normal kidney metabolic and homeostatic functions. Importantly, mice deficient for kidney Gcgr recapitulate some of the key pathophysiological features of chronic kidney disease.
Collapse
Affiliation(s)
- May-Yun Wang
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shangang Zhao
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Sam and Ann Barshop Institute for Longevity and Aging Studies, Division of Endocrinology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Toshiharu Onodera
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xue-Nan Sun
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qingzhang Zhu
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chao Li
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Na Li
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Megan Paredes
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laurent Gautron
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Denise K Marciano
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel J Drucker
- Lunenfeld-TanenbaumResearchInstitute, Mt. Sinai Hospital, Toronto, ON M5G1X5, Canada; Department of Medicine, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
14
|
Marstrand-Jørgensen AB, Sembach FE, Bak ST, Ougaard M, Christensen-Dalsgaard M, Rønn Madsen M, Jensen DM, Secher T, Heimbürger SMN, Fink LN, Hansen D, Hansen HH, Østergaard MV, Christensen M, Dalbøge LS. Shared and Distinct Renal Transcriptome Signatures in 3 Standard Mouse Models of Chronic Kidney Disease. Nephron Clin Pract 2024; 148:487-502. [PMID: 38354720 DOI: 10.1159/000535918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/04/2023] [Indexed: 02/16/2024] Open
Abstract
INTRODUCTION Several mouse models with diverse disease etiologies are used in preclinical research for chronic kidney disease (CKD). Here, we performed a head-to-head comparison of renal transcriptome signatures in standard mouse models of CKD to assess shared and distinct molecular changes in three mouse models commonly employed in preclinical CKD research and drug discovery. METHODS All experiments were conducted on male C57BL/6J mice. Mice underwent sham, unilateral ureter obstruction (UUO), or unilateral ischemic-reperfusion injury (uIRI) surgery and were terminated two- and 6-weeks post-surgery, respectively. The adenine-supplemented diet-induced (ADI) model of CKD was established by feeding with adenine diet for 6 weeks and compared to control diet feeding. For all models, endpoints included plasma biochemistry, kidney histology, and RNA sequencing. RESULTS All models displayed increased macrophage infiltration (F4/80 IHC) and fibrosis (collagen 1a1 IHC). Compared to corresponding controls, all models were characterized by an extensive number of renal differentially expressed genes (≥11,000), with a notable overlap in transcriptomic signatures across models. Gene expression markers of fibrosis, inflammation, and kidney injury supported histological findings. Interestingly, model-specific transcriptome signatures included several genes representing current drug targets for CKD, emphasizing advantages and limitations of the three CKD models in preclinical target and drug discovery. CONCLUSION The UUO, uIRI, and ADI mouse models of CKD have significant commonalities in their renal global transcriptome profile. Model-specific renal transcriptional signatures should be considered when selecting the specific model in preclinical target and drug discovery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Thomas Secher
- Gubra A/S, Hørsholm, Denmark
- Cell Imaging and Pharmacology, Cell Therapy R&D, Novo Nordisk A/S, Måløv, Denmark
| | | | - Lisbeth N Fink
- Gubra A/S, Hørsholm, Denmark
- Biotherapeutics Screening, Ferring Pharmaceuticals A/S, Kastrup, Denmark
| | - Ditte Hansen
- Department of Nephrology, Herlev-Gentofte Hospital, University of Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
15
|
Xue R, Wang Y, Geng L, Xiao H, Kumar V, Lan X, Malhotra A, Singhal PC, Chen J. Comprehensive analysis of the gene expression profile of the male and female BTBR mice with diabetic nephropathy. Int J Biol Macromol 2024; 257:128720. [PMID: 38101684 DOI: 10.1016/j.ijbiomac.2023.128720] [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: 07/23/2023] [Revised: 09/26/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
Comprehensive insight into the gender-based gene expression-related omics data in a rodent model of diabetic nephropathy (DN) is scarce. In the present study, the gender-based genes regulating different pathways involved in the progression of DN were explored through an unbiased RNA sequence of kidneys from BTBR mice with DN. We identified 17,739 and 17,981 genes in male and female DN mice; 1121 and 655 genes were expressed differentially (DEGs, differentially expressed genes) in male and female DN mice; both genders displayed only 195 DEGs. In the male DN mice, the number of upregulated genes was nearly the same as that of the down-regulated genes. In contrast, the number of upregulated genes was lesser than that of the down-regulated genes in the female DN mice, manifesting a remarkable gender disparity during the progression of DN in this animal model. Gene Ontology (GO) and KEGG-enriched results showed that most of these DEGs were related to the critical biological processes, including metabolic pathways, natural oxidation, bile secretion, and PPAR signaling; all are highly associated with DN. Notably, the DEGs significantly enriched for steroid hormone biosynthesis pathway were identified in both genders; the number of DEGs increased was 22 in male DN mice and 14 in female DN mice. Specifically, the Ugt1a10, Akr1c12, and Akr1c14 were upregulated in both genders. Interestingly, the Hsd11b1 gene was upregulated in female DN mice but downregulated in male DN mice. These results suggest that a significant gender-based variance in the gene expression occurs during the progression of DN and may be playing a role in the advancement of DN in the BTBR mouse model.
Collapse
Affiliation(s)
- Rui Xue
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Ying Wang
- Department of Pathogenic Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Lei Geng
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Haiting Xiao
- Key Laboratory of Luzhou City for Aging Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Vinod Kumar
- Department of Dermatology, Postgraduate Institute for Medical Education and Research, Chandigarh 160012, India
| | - Xiqian Lan
- Key Laboratory of Luzhou City for Aging Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ashwani Malhotra
- Feinstein Institute for Medical Research and Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, United States
| | - Pravin C Singhal
- Feinstein Institute for Medical Research and Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, United States.
| | - Jianning Chen
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
16
|
Nørregaard R, Mutsaers HAM, Frøkiær J, Kwon TH. Obstructive nephropathy and molecular pathophysiology of renal interstitial fibrosis. Physiol Rev 2023; 103:2827-2872. [PMID: 37440209 PMCID: PMC10642920 DOI: 10.1152/physrev.00027.2022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023] Open
Abstract
The kidneys play a key role in maintaining total body homeostasis. The complexity of this task is reflected in the unique architecture of the organ. Ureteral obstruction greatly affects renal physiology by altering hemodynamics, changing glomerular filtration and renal metabolism, and inducing architectural malformations of the kidney parenchyma, most importantly renal fibrosis. Persisting pathological changes lead to chronic kidney disease, which currently affects ∼10% of the global population and is one of the major causes of death worldwide. Studies on the consequences of ureteral obstruction date back to the 1800s. Even today, experimental unilateral ureteral obstruction (UUO) remains the standard model for tubulointerstitial fibrosis. However, the model has certain limitations when it comes to studying tubular injury and repair, as well as a limited potential for human translation. Nevertheless, ureteral obstruction has provided the scientific community with a wealth of knowledge on renal (patho)physiology. With the introduction of advanced omics techniques, the classical UUO model has remained relevant to this day and has been instrumental in understanding renal fibrosis at the molecular, genomic, and cellular levels. This review details key concepts and recent advances in the understanding of obstructive nephropathy, highlighting the pathophysiological hallmarks responsible for the functional and architectural changes induced by ureteral obstruction, with a special emphasis on renal fibrosis.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jørgen Frøkiær
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
17
|
Han X, Guo J, Wang M, Zhang N, Ren J, Yang Y, Chi X, Chen Y, Yao H, Zhao YL, Yang YG, Sun Y, Xu J. Dynamic DNA 5-hydroxylmethylcytosine and RNA 5-methycytosine Reprogramming During Early Human Development. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:805-822. [PMID: 35644351 PMCID: PMC10787118 DOI: 10.1016/j.gpb.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/18/2022] [Accepted: 05/05/2022] [Indexed: 06/15/2023]
Abstract
After implantation, complex and highly specialized molecular events render functionally distinct organ formation, whereas how the epigenome shapes organ-specific development remains to be fully elucidated. Here, nano-hmC-Seal, RNA bisulfite sequencing (RNA-BisSeq), and RNA sequencing (RNA-Seq) were performed, and the first multilayer landscapes of DNA 5-hydroxymethylcytosine (5hmC) and RNA 5-methylcytosine (m5C) epigenomes were obtained in the heart, kidney, liver, and lung of the human foetuses at 13-28 weeks with 123 samples in total. We identified 70,091 and 503 organ- and stage-specific differentially hydroxymethylated regions (DhMRs) and m5C-modified mRNAs, respectively. The key transcription factors (TFs), T-box transcription factor 20 (TBX20), paired box 8 (PAX8), krueppel-like factor 1 (KLF1), transcription factor 21 (TCF21), and CCAAT enhancer binding protein beta (CEBPB), specifically contribute to the formation of distinct organs at different stages. Additionally, 5hmC-enriched Alu elements may participate in the regulation of expression of TF-targeted genes. Our integrated studies reveal a putative essential link between DNA modification and RNA methylation, and illustrate the epigenetic maps during human foetal organogenesis, which provide a foundation for for an in-depth understanding of the epigenetic mechanisms underlying early development and birth defects.
Collapse
Affiliation(s)
- Xiao Han
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jia Guo
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mengke Wang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nan Zhang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jie Ren
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Ying Yang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Xu Chi
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Yusheng Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Yao
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yong-Liang Zhao
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yun-Gui Yang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yingpu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Jiawei Xu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
18
|
Sun Y, Ge J, Shao F, Ren Z, Huang Z, Ding Z, Dong L, Chen J, Zhang J, Zang Y. Long noncoding RNA AI662270 promotes kidney fibrosis through enhancing METTL3-mediated m 6 A modification of CTGF mRNA. FASEB J 2023; 37:e23071. [PMID: 37389924 DOI: 10.1096/fj.202202012rrr] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
The sustained release of profibrotic cytokines, mainly transforming growth factor-β (TGF-β), leads to the occurrence of kidney fibrosis and chronic kidney disease (CKD). Connective tissue growth factor (CTGF) appears to be an alternative target to TGF-β for antifibrotic therapy in CKD. In this study, we found that long noncoding RNA AI662270 was significantly increased in various renal fibrosis models. In vivo, ectopic expression of AI662270 alone was sufficient to activate interstitial fibroblasts and drive kidney fibrosis, whereas inhibition of AI662270 blocked the activation of interstitial fibroblasts and ameliorated kidney fibrosis in various murine models. Mechanistic studies revealed that overexpression of AI662270 significantly increased CTGF product, which was required for the role of AI662270 in driving kidney fibrosis. Furthermore, AI662270 binds to the CTGF promoter and directly interacts with METTL3, the methyltransferase of RNA N6 -methyladenosine (m6 A) modification. Functionally, AI662270-mediated recruitment of METTL3 increased the m6 A methylation of CTGF mRNA and consequently enhanced CTGF mRNA stability. In conclusion, our results support that AI662270 promotes CTGF expression at the posttranscriptional stage by recruiting METTL3 to the CTGF promoter and depositing m6 A modifications on the nascent mRNA, thereby, uncovering a novel regulatory mechanism of CTGF in the pathogenesis of kidney fibrosis.
Collapse
Affiliation(s)
- Yanyan Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Jia Ge
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Fang Shao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Zhengrong Ren
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Zhi Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
- State Key Laboratory of Analytical Chemistry for Life Sciences and Collaborative Innovation Center of Chemistry for Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| | - Yuhui Zang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P.R. China
| |
Collapse
|
19
|
Lindquist JA, Bernhardt A, Reichardt C, Sauter E, Brandt S, Rana R, Lindenmeyer MT, Philipsen L, Isermann B, Zhu C, Mertens PR. Cold Shock Domain Protein DbpA Orchestrates Tubular Cell Damage and Interstitial Fibrosis in Inflammatory Kidney Disease. Cells 2023; 12:1426. [PMID: 37408260 DOI: 10.3390/cells12101426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
DNA-binding protein A (DbpA) belongs to the Y-box family of cold shock domain proteins that exert transcriptional and translational activities in the cell via their ability to bind and regulate mRNA. To investigate the role of DbpA in kidney disease, we utilized the murine unilateral ureter obstruction (UUO) model, which recapitulates many features of obstructive nephropathy seen in humans. We observed that DbpA protein expression is induced within the renal interstitium following disease induction. Compared with wild-type animals, obstructed kidneys from Ybx3-deficient mice are protected from tissue injury, with a significant reduction in the number of infiltrating immune cells as well as in extracellular matrix deposition. RNAseq data from UUO kidneys show that Ybx3 is expressed by activated fibroblasts, which reside within the renal interstitium. Our data support a role for DbpA in orchestrating renal fibrosis and suggest that strategies targeting DbpA may be a therapeutic option to slow disease progression.
Collapse
Affiliation(s)
- Jonathan A Lindquist
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Anja Bernhardt
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Charlotte Reichardt
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Eva Sauter
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Sabine Brandt
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Rajiv Rana
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, 04103 Leipzig, Germany
| | - Maja T Lindenmeyer
- Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lars Philipsen
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig University, 04103 Leipzig, Germany
| | - Cheng Zhu
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou 310058, China
| | - Peter R Mertens
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
20
|
Chung YH, Huang GK, Kang CH, Cheng YT, Kao YH, Chien YS. MicroRNA-26a-5p Restoration Ameliorates Unilateral Ureteral Obstruction-Induced Renal Fibrosis In Mice Through Modulating TGF-β Signaling. J Transl Med 2023; 103:100131. [PMID: 36948295 DOI: 10.1016/j.labinv.2023.100131] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 02/14/2023] [Accepted: 03/04/2023] [Indexed: 03/24/2023] Open
Abstract
Renal fibrosis is a hallmark of chronic and progressive renal diseases characterized by excessive fibroblast proliferation, extracellular matrix accumulation, and loss of renal function, eventually leading to end-stage renal diseases. MicroRNA-26a-5p downregulation has been previously noted in the sera of unilateral ureteral occlusion (UUO)-injured mice, and exosome-mediated miR-26a-5p reportedly attenuated experimental pulmonary and cardiac fibrosis. This study evaluated the expression patterns of miR-26a in human tissue microarray with kidney fibrosis and in tissues from a mouse model of UUO-induced renal fibrosis. Histological analyses showed that miR-26a-5p was downregulated in human and mouse tissues with renal interstitial nephritis and fibrosis. Moreover, miR-26a-5p restoration by intravenous injection of a mimic agent prominently suppressed the expression of TGF-β1 and its cognate receptors, the inflammatory transcription factor NF-κB, epithelial-mesenchymal transition, and inflammatory markers in UUO-injured kidney tissues. In vitro miR-26a-5p mimic delivery significantly inhibited TGF-β1-induced activation of cultured rat kidney NRK-49F cells, in terms of downregulation of TGF-β1 receptors, restoration of epithelial marker E-cadherin, and suppression of mesenchymal markers, including vimentin, fibronectin, and α-smooth muscle actin, as well as TGF-β1/SMAD3 signaling activity. Our findings identified miR-26a-5p downregulation in kidney tissues from human interstitial nephritis and UUO-induced mouse kidney fibrosis. MiR-26a-5p restoration may exhibit an anti-fibrotic effect through the blockade of both TGF-β and NF-κB signaling axes and is considered a novel therapeutic target for treating obstruction-induced renal fibrosis.
Collapse
Affiliation(s)
- Yueh-Hua Chung
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Gong-Kai Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chih-Hsiung Kang
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Yuan-Tso Cheng
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Ying-Hsien Kao
- Department of Medical Research, E-Da Hospital, Kaohsiung 82445, Taiwan.
| | - Yu-Shu Chien
- Division of Nephrology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| |
Collapse
|
21
|
Hong Y, Zhang Y, Chen H, Tang X, Zhao H, Meng Z, Jia X, Liu W, Li X, Wang L, Zhong X, Bai X, Sun H, Kopylov P, Afina B, Shchekochikhin D, Zhang Y, Liu X, Fan Y. Genetic dissection of the impact of lncRNA AI662270 during the development of atherosclerosis. J Transl Med 2023; 21:97. [PMID: 36755320 PMCID: PMC9906833 DOI: 10.1186/s12967-023-03962-6] [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: 05/12/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Atherosclerosis is driven by synergistic interactions between pathological biomechanical and lipid metabolic factors. Long noncoding RNAs (LncRNAs) have been implicated in atherogenesis. The purpose of this study was to investigate the potential mechanism of lncRNA AI662270 on macrophage cholesterol transport in atherosclerosis. METHODS Apolipoprotein E deficiency (ApoE-/-) mice were fed a high fat diet for 16 weeks to construct atherosclerotic model, and the mice were injected with recombinant lentivirus carrying AI662270 gene to overexpress AI662270. Macrophages were cleared by liposomal clondronate in vivo. Fundamental experiments and functional assays, hematoxylin and eosin staining, oil red O staining and others, were performed to evaluate the function of AI662270 on atherogenesis. Peritoneal macrophages were treated with oxidized low density lipoprotein (ox-LDL) to simulate in vitro model. Mechanism assays, RNA-interacting protein immunoprecipitation, RNA-protein pulldown and others, were performed to study the regulatory mechanism of AI662270 in macrophages. RESULTS The novel AI662270 was mainly enriched in macrophages, but not in endothelial cells, smooth muscle cells and fibroblasts of mouse atherosclerotic lesions and was upregulated by ox-LDL. Overexpression of AI662270 resulted in lipid accumulation, larger atherosclerotic plaques and cardiac dysfunction in vivo. After macrophages were removed, the pro-atherogenic effect of AI662270 disappeared. Downregulation of AI662270 in macrophages protected against foam cell formation by potentiating cholesterol efflux and reducing intracellular total cholesterol. The opposite effect was observed in macrophage-specific AI662270-overexpressed cells in vitro. AI662270 bound to adenosine triphosphate-binding cassette transporter A1 (Abca1) responsible for regulating cholesterol efflux in macrophages. Forced expression of AI662270 in macrophages decreased Abca1 expression. The reverse occurred when expression of AI662270 was repressed. CONCLUSION These findings reveal an essential role for AI662270 in atherosclerosis progression by regulating cholesterol efflux from macrophages.
Collapse
Affiliation(s)
- Yang Hong
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Yue Zhang
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Hui Chen
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Xueqing Tang
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Hongrui Zhao
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Ziyu Meng
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Xueling Jia
- grid.410736.70000 0001 2204 9268Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319 Heilongjiang People’s Republic of China
| | - Wenfeng Liu
- grid.410736.70000 0001 2204 9268Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319 Heilongjiang People’s Republic of China
| | - Xiaohan Li
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Lin Wang
- grid.410736.70000 0001 2204 9268Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319 Heilongjiang People’s Republic of China
| | - Xinrui Zhong
- grid.410736.70000 0001 2204 9268Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319 Heilongjiang People’s Republic of China
| | - Xuefeng Bai
- grid.410736.70000 0001 2204 9268School of Medical Informatics, Harbin Medical University, Daqing Campus, Daqing, 163319 People’s Republic of China
| | - Heyang Sun
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Philipp Kopylov
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China ,grid.448878.f0000 0001 2288 8774Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Bestavashvili Afina
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China ,grid.448878.f0000 0001 2288 8774Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosofsky, I. M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Dmitry Shchekochikhin
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China ,grid.448878.f0000 0001 2288 8774Department of Preventive and Emergency Cardiology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Yong Zhang
- grid.410736.70000 0001 2204 9268Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081 Heilongjiang People’s Republic of China
| | - Xin Liu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China.
| | - Yuhua Fan
- Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319, Heilongjiang, People's Republic of China. .,Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China.
| |
Collapse
|
22
|
Reichelt-Wurm S, Pregler M, Wirtz T, Kretz M, Holler K, Banas B, Banas MC. The Interplay of NEAT1 and miR-339-5p Influences on Mesangial Gene Expression and Function in Various Diabetic-Associated Injury Models. Noncoding RNA 2022; 8:ncrna8040052. [PMID: 35893235 PMCID: PMC9326603 DOI: 10.3390/ncrna8040052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Mesangial cells (MCs), substantial cells for architecture and function of the glomerular tuft, take a key role in progression of diabetic kidney disease (DKD). Despite long standing researches and the need for novel therapies, the underlying regulatory mechanisms in MCs are elusive. This applies in particular to long non-coding RNAs (lncRNA) but also microRNAs (miRNAs). In this study, we investigated the expression of nuclear paraspeckle assembly transcript 1 (NEAT1), a highly conserved lncRNA, in several diabetes in-vitro models using human MCs. These cells were treated with high glucose, TGFβ, TNAα, thapsigargin, or tunicamycin. We analyzed the implication of NEAT1 silencing on mesangial cell migration, proliferation, and cell size as well as on mRNA and miRNA expression. Here, the miRNA hsa-miR-339-5p was not only identified as a potential interaction partner for NEAT1 but also for several coding genes. Furthermore, overexpression of hsa-miR-339-5p leads to a MC phenotype comparable to a NEAT1 knockdown. In-silico analyses also underline a relevant role of NEAT1 and hsa-miR-339-5p in mesangial physiology, especially in the context of DKD.
Collapse
Affiliation(s)
- Simone Reichelt-Wurm
- Department of Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany; (M.P.); (T.W.); (K.H.); (B.B.); (M.C.B.)
- Correspondence: ; Tel.: +49-941-944-7388
| | - Matthias Pregler
- Department of Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany; (M.P.); (T.W.); (K.H.); (B.B.); (M.C.B.)
| | - Tobias Wirtz
- Department of Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany; (M.P.); (T.W.); (K.H.); (B.B.); (M.C.B.)
| | - Markus Kretz
- Regensburg Center for Biochemistry (RCB), University of Regensburg, 93053 Regensburg, Germany;
| | - Kathrin Holler
- Department of Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany; (M.P.); (T.W.); (K.H.); (B.B.); (M.C.B.)
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany; (M.P.); (T.W.); (K.H.); (B.B.); (M.C.B.)
| | - Miriam C. Banas
- Department of Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany; (M.P.); (T.W.); (K.H.); (B.B.); (M.C.B.)
| |
Collapse
|
23
|
Zhang Y, Wang Y, Zheng G, Liu Y, Li J, Huang H, Xu C, Zeng Y, Zhang X, Qin J, Dai C, Hambrock HO, Hartmann U, Feng B, Mak KK, Liu Y, Lan HY, Huang Y, Zheng ZH, Xia Y. Follistatin-like 1 (FSTL1) interacts with Wnt ligands and Frizzled receptors to enhance Wnt/β-catenin signaling in obstructed kidneys in vivo. J Biol Chem 2022; 298:102010. [PMID: 35525270 PMCID: PMC9234244 DOI: 10.1016/j.jbc.2022.102010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
Follistatin (FS)-like 1 (FSTL1) is a member of the FS-SPARC (secreted protein, acidic and rich in cysteine) family of secreted and extracellular matrix proteins. The functions of FSTL1 have been studied in heart and lung injury as well as in wound healing; however, the role of FSTL1 in the kidney is largely unknown. Here, we show using single-cell RNA-Seq that Fstl1 was enriched in stromal cells in obstructed mouse kidneys. In addition, immunofluorescence demonstrated that FSTL1 expression was induced in fibroblasts during kidney fibrogenesis in mice and human patients. We demonstrate that FSTL1 overexpression increased renal fibrosis and activated the Wnt/β-catenin signaling pathway, known to promote kidney fibrosis, but not the transforming growth factor β (TGF-β), Notch, Hedgehog, or Yes-associated protein (YAP) signaling pathways in obstructed mouse kidneys, whereas inhibition of FSTL1 lowered Wnt/β-catenin signaling. Importantly, we show that FSTL1 interacted with Wnt ligands and the Frizzled (FZD) receptors but not the coreceptor lipoprotein receptor-related protein 6 (LRP6). Specifically, we found FSTL1 interacted with Wnt3a through its extracellular calcium-binding (EC) domain and von Willebrand factor type C-like (VWC) domain, and with FZD4 through its EC domain. Furthermore, we show that FSTL1 increased the association of Wnt3a with FZD4 and promoted Wnt/β-catenin signaling and fibrogenesis. The EC domain interacting with both Wnt3a and FZD4 also enhanced Wnt3a signaling. Therefore, we conclude that FSTL1 is a novel extracellular enhancer of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yu Zhang
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yang Wang
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Guoxun Zheng
- iHuman Institute, Shanghai Tech University, Shanghai, China
| | - Yang Liu
- Department of Nephrology, Center of Nephrology and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jinhong Li
- Department of Nephrology, Center of Nephrology and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Huihui Huang
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Chunhua Xu
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yelin Zeng
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyi Zhang
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jinzhong Qin
- The Key Laboratory of Model Animal for Disease Study of Ministry of Education, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Chunsun Dai
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Harald O Hambrock
- Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Ursula Hartmann
- Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Bo Feng
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Kingston Kinglun Mak
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
| | - Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory for Immune and Genetic Kidney Disease, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, and The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Zhi-Hua Zheng
- Department of Nephrology, Center of Nephrology and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Yin Xia
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory for Immune and Genetic Kidney Disease, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, and The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
24
|
Brandli A, Khong FL, Kong RCK, Kelly DJ, Fletcher EL. Transcriptomic analysis of choroidal neovascularization reveals dysregulation of immune and fibrosis pathways that are attenuated by a novel anti-fibrotic treatment. Sci Rep 2022; 12:859. [PMID: 35039609 PMCID: PMC8764037 DOI: 10.1038/s41598-022-04845-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/15/2021] [Indexed: 02/05/2023] Open
Abstract
Neovascular AMD (nAMD) leads to vision loss and is a leading cause of visual impairment in the industrialised world. Current treatments that target blood vessel growth have not been able to treat subretinal fibrosis and nAMD patients continue to lose vision. The molecular mechanisms involved in the development of fibrotic lesions in nAMD are not well understood. The aim of this study was to further understand subretinal fibrosis in the laser photocoagulation model of choroidal neovascularization (CNV) by studying the whole transcriptome of the RPE/choroid following CNV and the application of an anti-fibrotic following CNV. Seven days after laser induced CNV, RPE and choroid tissue was separated and underwent RNAseq. Differential expression analysis and pathway analysis revealed an over representation of immune signalling and fibrotic associated pathways in CNV compared to control RPE/choroid tissue. Comparisons between the mouse CNV model to human CNV revealed an overlap in upregulated expression for immune genes (Ccl2, Ccl8 and Cxcl9) and extracellular matrix remodeling genes (Comp, Lrcc15, Fndc1 and Thbs2). Comparisons between the CNV model and other fibrosis models showed an overlap of over 60% of genes upregulated in either lung or kidney mouse models of fibrosis. Treatment of CNV using a novel cinnamoyl anthranilate anti-fibrotic (OCX063) in the laser induced CNV model was selected as this class of drugs have previously been shown to target fibrosis. CNV lesion leakage and fibrosis was found to be reduced using OCX063 and gene expression of genes within the TGF-beta signalling pathway. Our findings show the presence of fibrosis gene expression pathways present in the laser induced CNV mouse model and that anti-fibrotic treatments offer the potential to reduce subretinal fibrosis in AMD.
Collapse
Affiliation(s)
- Alice Brandli
- Department of Anatomy and Physiology, The University of Melbourne, Grattan St, Parkville, VIC, 3010, Australia
| | - Fay L Khong
- Department of Medicine, The University of Melbourne, St Vincent's Hospital, Fitzroy, VIC, 3065, Australia
- Occurx Pty Ltd, 31 Queen St, Melbourne, VIC, 3000, Australia
| | - Roy C K Kong
- Department of Medicine, The University of Melbourne, St Vincent's Hospital, Fitzroy, VIC, 3065, Australia
- Occurx Pty Ltd, 31 Queen St, Melbourne, VIC, 3000, Australia
| | - Darren J Kelly
- Department of Medicine, The University of Melbourne, St Vincent's Hospital, Fitzroy, VIC, 3065, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Grattan St, Parkville, VIC, 3010, Australia.
| |
Collapse
|
25
|
Zhou Y, Luo Z, Liao C, Cao R, Hussain Z, Wang J, Zhou Y, Chen T, Sun J, Huang Z, Liu B, Zhang X, Guan Y, Deng T. MHC class II in renal tubules plays an essential role in renal fibrosis. Cell Mol Immunol 2021; 18:2530-2540. [PMID: 34556823 PMCID: PMC8545940 DOI: 10.1038/s41423-021-00763-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/18/2021] [Indexed: 02/08/2023] Open
Abstract
Immunomodulation is considered a potential therapeutic approach for chronic kidney disease (CKD). Although it has been previously reported that CD4+ T cells contribute to the development of renal fibrosis, the role of MHC class II (MHCII) in the development of renal fibrosis remains largely unknown. The present study reports that the expression of MHCII molecules in renal cortical tubules is upregulated in mouse renal fibrosis models generated by unilateral ureter obstruction (UUO) and folic acid (FA). Proximal tubule epithelial cells (PTECs) are functional antigen-presenting cells that promote the proliferation of CD4+ T cells in an MHCII-dependent manner. PTECs from mice with renal fibrosis had a stronger ability to induce T cell proliferation and cytokine production than control cells. Global or renal tubule-specific ablation of H2-Ab1 significantly alleviated renal fibrosis following UUO or FA treatment. Renal expression of profibrotic genes showed a consistent reduction in H2-Ab1 gene-deficient mouse lines. Moreover, there was a marked increase in renal tissue CD4+ T cells after UUO or FA treatment and a significant decrease following renal tubule-specific ablation of H2-Ab1. Furthermore, renal tubule-specific H2-Ab1 gene knockout mice exhibited higher proportions of regulatory T cells (Tregs) and lower proportions of Th2 cells in the UUO- or FA-treated kidneys. Finally, Immunohistochemistry (IHC) studies showed increased renal expression of MHCII and the profibrotic gene α smooth muscle actin (α-SMA) in CKD patients. Together, our human and mouse data demonstrate that renal tubular MHCII plays an important role in the pathogenesis of renal fibrosis.
Collapse
Affiliation(s)
- Yunfeng Zhou
- grid.263488.30000 0001 0472 9649Department of Physiology, Medical Research Center, Shenzhen University, Shenzhen, China
| | - Zhaokang Luo
- grid.263488.30000 0001 0472 9649Department of Physiology, Medical Research Center, Shenzhen University, Shenzhen, China
| | - Chenghui Liao
- grid.263488.30000 0001 0472 9649Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University, Shenzhen, China
| | - Rong Cao
- grid.263488.30000 0001 0472 9649Department of Nephrology, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zain Hussain
- grid.416992.10000 0001 2179 3554Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX USA
| | - Jie Wang
- Department of Internal Medicine, Shenzhen Guangming Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Yeting Zhou
- grid.263488.30000 0001 0472 9649School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Tie Chen
- grid.263488.30000 0001 0472 9649School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Jie Sun
- grid.263488.30000 0001 0472 9649Department of Biochemistry and Molecular Biology, Medical Research Center, Shenzhen University, Shenzhen, China
| | - Zhong Huang
- grid.263488.30000 0001 0472 9649Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University, Shenzhen, China
| | - Baohua Liu
- grid.263488.30000 0001 0472 9649Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Medical Research Center, Shenzhen University, Shenzhen, China
| | - Xiaoyan Zhang
- grid.411971.b0000 0000 9558 1426Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China
| | - Youfei Guan
- grid.411971.b0000 0000 9558 1426Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China
| | - Tuo Deng
- grid.452708.c0000 0004 1803 0208National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China ,Key Laboratory of Diabetes Immunology, Ministry of Education, Changsha, China ,grid.216417.70000 0001 0379 7164Metabolic Syndrome Research Center, Clinical Immunology Center, Central South University, Changsha, China
| |
Collapse
|
26
|
Huang J, Zhang Z, Liu B, Gao Y, Nie J, Wen S, Lai X, Liang H. Identification of circular RNA expression profiles in renal fibrosis induced by obstructive injury. Ren Fail 2021; 43:1368-1377. [PMID: 34602010 PMCID: PMC8491726 DOI: 10.1080/0886022x.2021.1979040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Advancing renal fibrosis is the common histopathological feature of chronic obstructive nephropathy, representing the final pathway of nearly all chronic and progressive nephropathies. Increasing evidences suggest that circular RNAs (circRNAs) are crucial regulatory molecules present at virtually every level of the cellular pathophysiological process. Nonetheless, there are a few evidences for the role of circRNAs in renal fibrosis induced by obstructive nephropathy. AIMS We performed RNA-seq analysis to analyze the expression profiles of circRNAs in the obstructed kidneys to identify the potential circRNAs and their network. METHODS With silk ligated the left ureter to establish a mice unilateral ureteral obstruction (UUO) model. Renal tissue circRNAs were obtained and were screened by a circRNA microarray. The circRNA-miRNA-mRNA regulatory network and the target genes were visualized using Cytoscape software. RESULTS The microarray results showed that 5454 and 2935 circRNAs were detected in the control and UUO group, respectively. There were 605 circRNAs up-regulated and 745 circRNAs down-regulated in the obstructive kidneys. The top 5 up-regulated and down-regulated circRNAs were chosen for predicting the circRNA/miRNA/target mRNAs triple network. The GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis showed that these circRNAs and the triple network were enriched in the process of apoptosis, p53 signaling pathway, cell growth and cell death, which might participate in the pathogenesis of obstructive nephrology. CONCLUSION Our results show that the dis-regulated circRNAs might play crucial roles in the pathogenesis of obstructive nephropathy, which proceeds to identify novel therapeutic targets for chronic kidney disease.
Collapse
Affiliation(s)
- Jiangju Huang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China.,Department of Anesthesiology, The First Hospital of ChangSha, ChangSha, China
| | - Zhihao Zhang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Benquan Liu
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Ying Gao
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Jiayi Nie
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Shihong Wen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaohong Lai
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Hua Liang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
27
|
Zhang W, Xu H, Qiao R, Zhong B, Zhang X, Gu J, Zhang X, Wei L, Wang X. ARIC: accurate and robust inference of cell type proportions from bulk gene expression or DNA methylation data. Brief Bioinform 2021; 23:6361035. [PMID: 34472588 DOI: 10.1093/bib/bbab362] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 11/12/2022] Open
Abstract
Quantifying cell proportions, especially for rare cell types in some scenarios, is of great value in tracking signals associated with certain phenotypes or diseases. Although some methods have been proposed to infer cell proportions from multicomponent bulk data, they are substantially less effective for estimating the proportions of rare cell types which are highly sensitive to feature outliers and collinearity. Here we proposed a new deconvolution algorithm named ARIC to estimate cell type proportions from gene expression or DNA methylation data. ARIC employs a novel two-step marker selection strategy, including collinear feature elimination based on the component-wise condition number and adaptive removal of outlier markers. This strategy can systematically obtain effective markers for weighted $\upsilon$-support vector regression to ensure a robust and precise rare proportion prediction. We showed that ARIC can accurately estimate fractions in both DNA methylation and gene expression data from different experiments. We further applied ARIC to the survival prediction of ovarian cancer and the condition monitoring of chronic kidney disease, and the results demonstrate the high accuracy and robustness as well as clinical potentials of ARIC. Taken together, ARIC is a promising tool to solve the deconvolution problem of bulk data where rare components are of vital importance.
Collapse
Affiliation(s)
- Wei Zhang
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| | - Hanwen Xu
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| | - Rong Qiao
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| | - Bixi Zhong
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xianglin Zhang
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jin Gu
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xuegong Zhang
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| | - Lei Wei
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xiaowo Wang
- Ministry of Education Key Laboratory of Bioinformatics; Center for Synthetic and Systems Biology; Bioinformatics Division, Beijing National Research Center for Information Science and Technology; Department of Automation, Tsinghua University, Beijing 100084, China
| |
Collapse
|
28
|
Higgins CE, Tang J, Higgins SP, Gifford CC, Mian BM, Jones DM, Zhang W, Costello A, Conti DJ, Samarakoon R, Higgins PJ. The Genomic Response to TGF-β1 Dictates Failed Repair and Progression of Fibrotic Disease in the Obstructed Kidney. Front Cell Dev Biol 2021; 9:678524. [PMID: 34277620 PMCID: PMC8284093 DOI: 10.3389/fcell.2021.678524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Tubulointerstitial fibrosis is a common and diagnostic hallmark of a spectrum of chronic renal disorders. While the etiology varies as to the causative nature of the underlying pathology, persistent TGF-β1 signaling drives the relentless progression of renal fibrotic disease. TGF-β1 orchestrates the multifaceted program of kidney fibrogenesis involving proximal tubular dysfunction, failed epithelial recovery or re-differentiation, capillary collapse and subsequent interstitial fibrosis eventually leading to chronic and ultimately end-stage disease. An increasing complement of non-canonical elements function as co-factors in TGF-β1 signaling. p53 is a particularly prominent transcriptional co-regulator of several TGF-β1 fibrotic-response genes by complexing with TGF-β1 receptor-activated SMADs. This cooperative p53/TGF-β1 genomic cluster includes genes involved in cellular proliferative control, survival, apoptosis, senescence, and ECM remodeling. While the molecular basis for this co-dependency remains to be determined, a subset of TGF-β1-regulated genes possess both p53- and SMAD-binding motifs. Increases in p53 expression and phosphorylation, moreover, are evident in various forms of renal injury as well as kidney allograft rejection. Targeted reduction of p53 levels by pharmacologic and genetic approaches attenuates expression of the involved genes and mitigates the fibrotic response confirming a key role for p53 in renal disorders. This review focuses on mechanisms underlying TGF-β1-induced renal fibrosis largely in the context of ureteral obstruction, which mimics the pathophysiology of pediatric unilateral ureteropelvic junction obstruction, and the role of p53 as a transcriptional regulator within the TGF-β1 repertoire of fibrosis-promoting genes.
Collapse
Affiliation(s)
- Craig E. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Jiaqi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Stephen P. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Cody C. Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Badar M. Mian
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - David M. Jones
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, NY, United States
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Angelica Costello
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - David J. Conti
- Division of Transplantation Surgery, Department of Surgery, Albany Medical College, Albany, NY, United States
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Paul J. Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
- The Urological Institute of Northeastern New York, Albany, NY, United States
- Division of Urology, Department of Surgery, Albany Medical College, Albany, NY, United States
| |
Collapse
|
29
|
Dolivo D, Rodrigues A, Sun L, Li Y, Hou C, Galiano R, Hong SJ, Mustoe T. The Na x (SCN7A) channel: an atypical regulator of tissue homeostasis and disease. Cell Mol Life Sci 2021; 78:5469-5488. [PMID: 34100980 PMCID: PMC11072345 DOI: 10.1007/s00018-021-03854-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/15/2021] [Accepted: 05/08/2021] [Indexed: 12/15/2022]
Abstract
Within an articulately characterized family of ion channels, the voltage-gated sodium channels, exists a black sheep, SCN7A (Nax). Nax, in contrast to members of its molecular family, has lost its voltage-gated character and instead rapidly evolved a new function as a concentration-dependent sensor of extracellular sodium ions and subsequent signal transducer. As it deviates fundamentally in function from the rest of its family, and since the bulk of the impressive body of literature elucidating the pathology and biochemistry of voltage-gated sodium channels has been performed in nervous tissue, reports of Nax expression and function have been sparse. Here, we investigate available reports surrounding expression and potential roles for Nax activity outside of nervous tissue. With these studies as justification, we propose that Nax likely acts as an early sensor that detects loss of tissue homeostasis through the pathological accumulation of extracellular sodium and/or through endothelin signaling. Sensation of homeostatic aberration via Nax then proceeds to induce pathological tissue phenotypes via promotion of pro-inflammatory and pro-fibrotic responses, induced through direct regulation of gene expression or through the generation of secondary signaling molecules, such as lactate, that can operate in an autocrine or paracrine fashion. We hope that our synthesis of much of the literature investigating this understudied protein will inspire more research into Nax not simply as a biochemical oddity, but also as a potential pathophysiological regulator and therapeutic target.
Collapse
Affiliation(s)
- David Dolivo
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Adrian Rodrigues
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Lauren Sun
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Yingxing Li
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Chun Hou
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
- Department of Plastic and Cosmetic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Robert Galiano
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Seok Jong Hong
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA.
- , 300 E. Superior St., Chicago, IL, 60611, USA.
| | - Thomas Mustoe
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA.
- , 737 N. Michigan Ave., Chicago, IL, 60611, USA.
| |
Collapse
|
30
|
Zhang X, Chen Q, Zhang L, Zheng H, Lin C, Yang Q, Liu T, Zhang H, Chen X, Ren L, Shan W. Tubule-specific protein nanocages potentiate targeted renal fibrosis therapy. J Nanobiotechnology 2021; 19:156. [PMID: 34039349 PMCID: PMC8157627 DOI: 10.1186/s12951-021-00900-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023] Open
Abstract
Background Despite the dramatic advances in modern medicine, efficient therapeutic measures for renal fibrosis remain limited. Celastrol (CLT) is effective in treating renal fibrosis in rat models, while causing severe systemic toxicity. Thus, we designed a tubule-specific nanocage (K3-HBc NCs) that effectively deliver CLT to tubular epithelial cell in a virus-like manner. The targeting ligand (K3) to tubular epithelial cells was displayed on the surface of Hepatitis B core protein (HBc) NCs by genetic fusion to the major immunodominant loop region. Ultra-small CLT nanodots were subtly encapsulated into the cavity through electrostatic interaction with the disassembly and reassembly of K3-HBc NCs, to yield K3-HBc/CLT complex. The efficacy of K3-HBc/CLT NCs were demonstrated in Unilateral ureteral obstruction (UUO)-induced renal fibrosis. Results The self-assembled K3-HBc/CLT could specifically target tubular epithelial cells via affinity with K3 ligand binding to the megalin receptor, significantly attenuating renal fibrosis. Remarkably, K3-HBc/CLT NCs significantly increased therapeutic efficacy and reduced the systemic toxicity in comparison with free CLT in UUO-induced mouse renal fibrosis model. Importantly, analysis of RNA sequencing data suggested that the anti-fibrotic effect of K3-HBc/CLT could be attributed to suppression of premature senescence in tubular epithelial cells via p21Cip1 and p16Ink4a pathway. Conclusion The tubule-specific K3-HBc/CLT represented a promising option to realize precise treatment for renal fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00900-w.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Qian Chen
- Biomedical Analysis Center, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Liyuan Zhang
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Haiping Zheng
- School of Medicine, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Chunjie Lin
- School of Life Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Qunfang Yang
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Tao Liu
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Haigang Zhang
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Xiaohong Chen
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Lei Ren
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, People's Republic of China.
| | - Wenjun Shan
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China. .,School of Medicine, Huaqiao University, Quanzhou, 362021, People's Republic of China.
| |
Collapse
|
31
|
Gu YY, Dou JY, Huang XR, Liu XS, Lan HY. Transforming Growth Factor-β and Long Non-coding RNA in Renal Inflammation and Fibrosis. Front Physiol 2021; 12:684236. [PMID: 34054586 PMCID: PMC8155637 DOI: 10.3389/fphys.2021.684236] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
Renal fibrosis is one of the most characterized pathological features in chronic kidney disease (CKD). Progressive fibrosis eventually leads to renal failure, leaving dialysis or allograft transplantation the only clinical option for CKD patients. Transforming growth factor-β (TGF-β) is the key mediator in renal fibrosis and is an essential regulator for renal inflammation. Therefore, the general blockade of the pro-fibrotic TGF-β may reduce fibrosis but may risk promoting renal inflammation and other side effects due to the diverse role of TGF-β in kidney diseases. Long non-coding RNAs (lncRNAs) are RNA transcripts with more than 200 nucleotides and have been regarded as promising therapeutic targets for many diseases. This review focuses on the importance of TGF-β and lncRNAs in renal inflammation, fibrogenesis, and the potential applications of TGF-β and lncRNAs as the therapeutic targets and biomarkers in renal fibrosis and CKD are highlighted.
Collapse
Affiliation(s)
- Yue-Yu Gu
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jing-Yun Dou
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Weihai Hospital of Traditional Chinese Medicine, Weihai, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Xu-Sheng Liu
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
32
|
Sun D, Xie N, Wang X, Wu W, Li X, Chen X, Qian G, Li C, Zhang H, Jiang Y, Ye D, Liu D, Hu Y, Wang J, Chen W, Zhao Q, Zeng M, Zhang J, Wang L, Zhang X. Serum RelB is correlated with renal fibrosis and predicts chronic kidney disease progression. Clin Transl Med 2021; 11:e362. [PMID: 34047463 PMCID: PMC8140188 DOI: 10.1002/ctm2.362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 11/10/2022] Open
Affiliation(s)
- Donglin Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Ningxia Xie
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Xi Wang
- Shenzhen Longhua District Central HospitalNephrology Department
| | - Wenquan Wu
- Southern Medical University Affiliated Longhua People's HospitalClinical Laboratory
| | - Xiu‐Yong Li
- NO.2 People's Hospital of Fuyang CityFuyang236015China
| | - Xiangqiu Chen
- Shenzhen HospitalSouthern Medical UniversityUrology Department
| | - Guojun Qian
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Cuifeng Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Haohao Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Yuhang Jiang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Deji Ye
- Shanghai Institute of Nutrition and HealthShanghai Institutes for Biological SciencesUniversity of Chinese Academy of SciencesChinese Academy of SciencesCAS Key Laboratory of Tissue Microenvironment and Tumor
| | - Dandan Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Yiming Hu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Jingyao Wang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Weifeng Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Qiumei Zhao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| | - Min Zeng
- Nephrology DepartmentSouthern Medical University Affiliated Longhua People's Hospital
| | - Junwei Zhang
- Nephrology DepartmentSouthern Medical University Affiliated Longhua People's Hospital
| | - Li Wang
- Nephrology DepartmentSouthern Medical University Affiliated Longhua People's Hospital
| | - Xiaoren Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityGuangzhou Municipal and Guangdong Provincial KeyLaboratory of Protein Modification and DegradationState Key Laboratory of Respiratory Disease
| |
Collapse
|
33
|
Su H, Xie J, Wen L, Wang S, Chen S, Li J, Qi C, Zhang Q, He X, Zheng L, Wang L. LncRNA Gas5 regulates Fn1 deposition via Creb5 in renal fibrosis. Epigenomics 2021; 13:699-713. [PMID: 33876672 DOI: 10.2217/epi-2020-0449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aim: Although studies on lncRNAs in renal fibrosis have focused on target genes and functions of lncRNAs, a comprehensive interaction analysis of lncRNAs is lacking. Materials & methods: Differentially expressed genes in renal fibrosis were screened, and the interaction between lncRNAs and miRNAs was searched. Results: We constructed a ceRNA network associated with renal fibrosis, by which we found the transcription factor Creb5, a target gene of lncRNA Gas5 that might regulate extracellular Fn1 deposition. Conclusion: Our study not only provides a theoretical basis for the ceRNA regulation mechanism of Gas5 but also provides experimental evidence supporting the use of Gas5 targeting in the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Huanhou Su
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Jingzhou Xie
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Lijing Wen
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Shunyi Wang
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Sishuo Chen
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Jiangchao Li
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Cuiling Qi
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Qianqian Zhang
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Xiaodong He
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Lingyun Zheng
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| | - Lijing Wang
- School of Life Sciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, P.R. China
| |
Collapse
|
34
|
Chou LF, Chen TW, Yang HY, Tian YC, Chang MY, Hung CC, Hsu SH, Tsai CY, Ko YC, Yang CW. Transcriptomic signatures of exacerbated progression in leptospirosis subclinical chronic kidney disease with secondary nephrotoxic injury. Am J Physiol Renal Physiol 2021; 320:F1001-F1018. [PMID: 33779314 DOI: 10.1152/ajprenal.00640.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
High-incidence regions of leptospirosis caused by Leptospira spp. coincide with chronic kidney disease. This study investigated whether asymptomatic leptospirosis is an emerging culprit that predisposes to progressive chronic kidney disease when superimposed on secondary nephrotoxic injury. Kidney histology/function and whole transcriptomic profiles were evaluated for Leptospira-infected C57/BL6 mice with adenine-induced kidney injury. The extent of tubulointerstitial kidney lesions and expression of inflammation/fibrosis genes in infected mice with low-dose (0.1%) adenine, particularly in high-dose (0.2%) adenine-fed superimposed on Leptospira-infected mice, were significantly increased compared with mice following infection or adenine diet alone, and the findings are consistent with renal transcriptome analysis. Pathway enrichment findings showed that integrin-β- and fibronectin-encoding genes had distinct expression within the integrin-linked kinase-signaling pathway, which were upregulated in 0.2% adenine-fed Leptospira-infected mice but not in 0.2% adenine-fed mice, indicating that background subclinical Leptospiral infection indeed enhanced subsequent secondary nephrotoxic kidney injury and potential pathogenic molecules associated with secondary nephrotoxic leptospirosis. Comparative analysis of gene expression patterns with unilateral ureteric obstruction-induced mouse renal fibrosis and patients with chronic kidney disease showed that differentially expressed orthologous genes such as hemoglobin-α2, PDZ-binding kinase, and DNA topoisomerase II-α were identified in infected mice fed with low-dose and high-dose adenine, respectively, revealing differentially expressed signatures identical to those found in the datasets and may serve as markers of aggravated kidney progression. This study indicates that background subclinical leptospirosis, when subjected to various degrees of subsequent secondary nephrotoxic injury, may predispose to exacerbated fibrosis, mimicking the pathophysiological process of progressive chronic kidney disease.NEW & NOTEWORTHY Leptospira-infected mice followed by secondary nephrotoxic injury exacerbated immune/inflammatory responses and renal fibrosis. Comparison with the murine model revealed candidates involved in the progression of renal fibrosis in chronic kidney disease (CKD). Comparative transcriptome study suggests that secondary nephrotoxic injury in Leptospira-infected mice recapitulates the gene expression signatures found in CKD patients. This study indicates that secondary nephrotoxic injury may exacerbate CKD in chronic Leptospira infection implicating in the progression of CKD of unknown etiology.
Collapse
Affiliation(s)
- Li-Fang Chou
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Ting-Wen Chen
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Huang-Yu Yang
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Chung Tian
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Yang Chang
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Chieh Hung
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shen-Hsing Hsu
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chung-Ying Tsai
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Yi-Ching Ko
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Wei Yang
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
35
|
Identification of candidate lncRNA biomarkers for renal fibrosis: A systematic review. Life Sci 2020; 262:118566. [PMID: 33038373 DOI: 10.1016/j.lfs.2020.118566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/09/2020] [Accepted: 10/01/2020] [Indexed: 12/21/2022]
Abstract
AIMS To combine the results of dysregulated lncRNAs in individual renal fibrosis lncRNA expression profiling studies and to identify potential lncRNA biomarkers. MATERIALS AND METHODS We systematically searched three databases to identify lncRNA expression studies of renal fibrosis in animal models and humans. The lncRNA expression data were extracted from 24 included studies, and a lncRNA vote-counting strategy was applied to identify significant lncRNA biomarkers. The lncLocator algorithm was utilized to predict the potential subcellular localization of these lncRNAs. The predicted targets of the identified lncRNA biomarkers were obtained by searching LncBase v.2 and catRAPID. Finally, GO enrichment and KEGG pathway analyses were performed. KEY FINDINGS We recognized a significant lncRNA signature of 95 differentially expressed lncRNAs in 731 samples from rodent models of renal fibrosis and CKD patients, among which TCONS_01181049 and TCONS_01496394 were commonly upregulated in both urine and renal tissues, while lncRNA-Cancer Susceptibility Candidate 2 was downregulated in both blood and renal tissues. About 73.33% dysregulated lncRNAs in renal fibrosis animal models and 81.82% dysregulated lncRNAs in CKD patients were predicted to be localized to the cytoplasm. The most relevant biological processes and molecular functions associated with these lncRNAs were mRNA processing and RNA binding. SIGNIFICANCE The present systematic review identified 95 significantly dysregulated lncRNAs from 24 studies and future investigations should focus on exploring their potential effects on renal fibrosis and their clinical utility as biomarkers or therapeutic targets.
Collapse
|
36
|
Chen Y, Ding Y, Wang LM. Tripartite motif-containing 35 (TRIM35) is up-regulated in UUO-induced renal fibrosis animal model. Histol Histopathol 2020; 35:1427-1435. [PMID: 32955098 DOI: 10.14670/hh-18-255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Renal fibrosis has been recognized as a serious health threat in the world because of the high cost of treatment and poor prognosis. However, the molecular mechanism of renal fibrosis is still largely unknown. In this study, we aimed at illustrating the role of TRIM35 in the renal fibrosis process. A UUO mouse model and a TGF-β1-induced tubulointerstitial fibrosis model were constructed for the research of renal fibrosis at animal and cell level, respectively. Hematoxylin-eosin and Masson staining were used for visualizing the pathological change. qRT-PCR, Western blot analysis and immunohistochemical staining were used to detect the expression of fibrosis-associated proteins and TRIM35. The results showed that, after the modeling, the expressions of α-SMA, Collagen I, Collagen III, Fibronectin and Snail1 were up-regulated, while the expression of E-cadherin was down-regulated, indicating the successful construction of animal and cell models. More importantly, TRIM35 was proved to be up-regulated in both animal and cell models. Therefore, this study demonstrates the potential promotional effect of TRIM35 in the renal fibrosis process, which may prove to be a new biomarker for the diagnosis and development of new treatments of renal fibrosis.
Collapse
Affiliation(s)
- Yu Chen
- Organ Transplantation Institute of PLA, Chang zheng Hospital, Naval Medical University, Shanghai, China
| | - Yue Ding
- Organ Transplantation Institute of PLA, Chang zheng Hospital, Naval Medical University, Shanghai, China
| | - Li-Ming Wang
- Organ Transplantation Institute of PLA, Chang zheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
37
|
Zhang Z, Wu W, Fang X, Lu M, Wu H, Gao C, Xia Z. Sox9 promotes renal tubular epithelial‑mesenchymal transition and extracellular matrix aggregation via the PI3K/AKT signaling pathway. Mol Med Rep 2020; 22:4017-4030. [PMID: 32901875 DOI: 10.3892/mmr.2020.11488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 06/05/2020] [Indexed: 11/05/2022] Open
Abstract
Sox9 is important for multiple aspects of development, such as testis, pancreas and heart development. Previous studies have reported that Sox9 induced epithelial‑mesenchymal transition (EMT) and extracellular matrix (ECM) production in organ fibrosis and associated diseases, such as vascular calcification. However, to the best of our knowledge, the role and underlying mechanism of action of Sox9 in renal fibrogenesis remains unknown. The results of the present study revealed that Sox9 expression levels were upregulated in the tubular epithelial cells of a rat model of obstructive nephropathy. Furthermore, the overexpression of Sox9 in NRK‑52E cells was discovered to promote renal tubular EMT and ECM aggregation, and these fibrogenic actions were potentiated by TGF‑β1. Notably, RNA‑sequencing analysis indicated the possible regulatory role of the PI3K/AKT signaling pathway in Sox9‑mediated renal tubular EMT and ECM aggregation. It was further demonstrated that the expression levels of phosphorylated AKT were upregulated in NRK‑52E cells overexpressing Sox9, while the PI3K inhibitors, LY29002 and wortmannin, inhibited the renal tubular EMT and ECM aggregation induced by the overexpression of Sox9 in NEK‑52E cells. In conclusion, the findings of the present study suggested that Sox9 may serve a profibrotic role in the development of renal tubular EMT and ECM aggregation via the PI3K/AKT signaling pathway. Therefore, Sox9 may be considered as a promising target for treating renal fibrosis.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- Department of Pediatrics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Wei Wu
- Department of Pediatrics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Xiang Fang
- Department of Pediatrics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Mei Lu
- Department of Pediatrics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Heyan Wu
- Department of Pediatrics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Chunlin Gao
- Department of Pediatrics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Zhengkun Xia
- Department of Pediatrics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
38
|
Brandt S, Ballhause TM, Bernhardt A, Becker A, Salaru D, Le-Deffge HM, Fehr A, Fu Y, Philipsen L, Djudjaj S, Müller AJ, Kramann R, Ibrahim M, Geffers R, Siebel C, Isermann B, Heidel FH, Lindquist JA, Mertens PR. Fibrosis and Immune Cell Infiltration Are Separate Events Regulated by Cell-Specific Receptor Notch3 Expression. J Am Soc Nephrol 2020; 31:2589-2608. [PMID: 32859670 DOI: 10.1681/asn.2019121289] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/12/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Kidney injuries that result in chronic inflammation initiate crosstalk between stressed resident cells and infiltrating immune cells. In animal models, whole-body receptor Notch3 deficiency protects from leukocyte infiltration and organ fibrosis. However, the relative contribution of Notch3 expression in tissue versus infiltrating immune cells is unknown. METHODS Chimeric mice deficient for Notch3 in hematopoietic cells and/or resident tissue cells were generated, and kidney fibrosis and inflammation after unilateral ureteral obstruction (UUO) were analyzed. Adoptive transfer of labeled bone marrow-derived cells validated the results in a murine Leishmania ear infection model. In vitro adhesion assays, integrin activation, and extracellular matrix production were analyzed. RESULTS Fibrosis follows UUO, but inflammatory cell infiltration mostly depends upon Notch3 expression in hematopoietic cells, which coincides with an enhanced proinflammatory milieu (e.g., CCL2 and CCL5 upregulation). Notch3 expression on CD45+ leukocytes plays a prominent role in efficient cell transmigration. Functionally, leukocyte adhesion and integrin activation are abrogated in the absence of receptor Notch3. Chimeric animal models also reveal that tubulointerstitial fibrosis develops, even in the absence of prominent leukocyte infiltrates after ureteral obstruction. Deleting Notch3 receptors on resident cells blunts kidney fibrosis, ablates NF-κB signaling, and lessens matrix deposition. CONCLUSIONS Cell-specific receptor Notch3 signaling independently orchestrates leukocyte infiltration and organ fibrosis. Interference with Notch3 signaling may present a novel therapeutic approach in inflammatory as well as fibrotic diseases.
Collapse
Affiliation(s)
- Sabine Brandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany
| | - Tobias M Ballhause
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anja Bernhardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany
| | - Annika Becker
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Delia Salaru
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Hien Minh Le-Deffge
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Alexander Fehr
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany
| | - Yan Fu
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Lars Philipsen
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sonja Djudjaj
- Institute of Pathology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Andreas J Müller
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Intravital Microscopy of Infection and Immunity Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Mahmoud Ibrahim
- Department of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Robert Geffers
- Genome Analytics Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Chris Siebel
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, California
| | - Berend Isermann
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany.,Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Florian H Heidel
- Department of Hematology and Oncology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Department of Internal Medicine II, Hematology and Oncology, Friedrich Schiller University Medical Center, Jena, Germany.,Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Jonathan A Lindquist
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany
| | - Peter R Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany .,Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
39
|
Konstantinou E, Zagoriti Z, Pyriochou A, Poulas K. Microcurrent Stimulation Triggers MAPK Signaling and TGF-β1 Release in Fibroblast and Osteoblast-Like Cell Lines. Cells 2020; 9:E1924. [PMID: 32825091 PMCID: PMC7564311 DOI: 10.3390/cells9091924] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/08/2020] [Accepted: 08/18/2020] [Indexed: 12/26/2022] Open
Abstract
Wound healing constitutes an essential process for all organisms and involves a sequence of three phases. The disruption or elongation of any of these phases can lead to a chronic or non-healing wound. Electrical stimulation accelerates wound healing by mimicking the current that is generated in the skin after any injury. Here, we sought to identify the molecular mechanisms involved in the healing process following in vitro microcurrent stimulation-a type of electrotherapy. Our results concluded that microcurrents promote cell proliferation and migration in an ERK 1/2- or p38-dependent way. Furthermore, microcurrents induce the secretion of transforming growth factor-beta-1 (TGF-β1) in fibroblasts and osteoblast-like cells. Interestingly, transcriptomic analysis uncovered that microcurrents enhance the transcriptional activation of genes implicated in Hedgehog, TGF-β1 and MAPK signaling pathways. Overall, our results demonstrate that microcurrents may enhance wound closure through a combination of signal transductions, via MAPK's phosphorylation, and the transcriptional activation of specific genes involved in the healing process. These mechanisms should be further examined in vivo, in order to verify the beneficial effects of microcurrents in wound or fracture healing.
Collapse
Affiliation(s)
| | | | | | - Konstantinos Poulas
- Laboratory of Molecular Biology and Immunology, Department of Pharmacy, University of Patras, 26504 Rio, Greece; (E.K.); (Z.Z.); (A.P.)
| |
Collapse
|
40
|
Das D, Mawlong GT, Sarki YN, Singh AK, Chikkaputtaiah C, Boruah HPD. Transcriptome analysis of crude oil degrading Pseudomonas aeruginosa strains for identification of potential genes involved in crude oil degradation. Gene 2020; 755:144909. [PMID: 32569720 DOI: 10.1016/j.gene.2020.144909] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/05/2020] [Accepted: 06/17/2020] [Indexed: 12/27/2022]
Abstract
In the microbial world, bacteria are the most effective agents in petroleum hydrocarbons (PHs) degradation, utilization/mineralization and they serve as essential degraders of crude oil contaminated environment. Some genes and traits are involved in the hydrocarbon utilization process for which transcriptome analyses are important to identify differentially expressed genes (DEGs) among different conditions, leading to a new understanding of genes or pathways associated with crude oil degradation. In this work, three crude oil utilizing Pseudomonas aeruginosa strains designated as N002, TP16 and J001 subjected to transcriptome analyses revealed a total of 81, 269 and 137 significant DEGs. Among them are 80 up-regulated genes and one downregulated gene of N002, 121 up- regulated and 148 down-regulated genes of TP16, 97 up-regulated and 40 down-regulated genes of J001 which are involved in various metabolic pathways. TP16 strain has shown more number of DEGs upon crude oil treatment in comparison to the other two strains. Through quantitative real time polymerase chain reaction (qRT-PCR), the selected DEGs of each strain from transcriptome data were substantiated. The results have shown that the up- regulated and down-regulated genes observed by qRT-PCR were consistent with transcriptome data. Taken together, our transcriptome results have revealed that TP16 is a potential P. aeruginosa strain for functional analysis of identified potential DEGs involved in crude oil degradation.
Collapse
Affiliation(s)
- Dhrubajyoti Das
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India
| | - Gabriella T Mawlong
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India
| | - Yogita N Sarki
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Innovative and Scientific Research (AcSIR), CSIR-NEIST, Jorhat 785006, Assam, India
| | - Anil Kumar Singh
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Innovative and Scientific Research (AcSIR), CSIR-NEIST, Jorhat 785006, Assam, India
| | - Channakeshavaiah Chikkaputtaiah
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Innovative and Scientific Research (AcSIR), CSIR-NEIST, Jorhat 785006, Assam, India
| | - Hari Prasanna Deka Boruah
- Biotechnology Group, Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat 785006, Assam, India; Academy of Innovative and Scientific Research (AcSIR), CSIR-NEIST, Jorhat 785006, Assam, India.
| |
Collapse
|
41
|
Liu P, Zhang B, Chen Z, He Y, Du Y, Liu Y, Chen X. m 6A-induced lncRNA MALAT1 aggravates renal fibrogenesis in obstructive nephropathy through the miR-145/FAK pathway. Aging (Albany NY) 2020; 12:5280-5299. [PMID: 32203053 PMCID: PMC7138587 DOI: 10.18632/aging.102950] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 03/09/2020] [Indexed: 01/15/2023]
Abstract
Renal fibrosis is a key factor in chronic kidney disease (CKD). Long non-coding RNAs (lncRNAs) play important roles in the physiological and pathological progression of human diseases. However, the roles and underlying mechanisms of lncRNAs in renal fibrosis still need to be discovered. In this study, we first displayed the increased lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) expression in renal fibrosis in patients with obstructive nephropathy (ON). Then we found that transforming growth factor beta 1 (TGF-β1) induced epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) protein deposition, which promoted the viability, proliferation and migration of human renal proximal tubular epithelial (HK2) cells. Next, MALAT1/miR-145/focal adhesion kinase (FAK) pathway was confirmed to play an importment role in TGF-β1-induced renal fibrosis. In addition, the MALAT1/miR-145/FAK pathway was involved in the effect of dihydroartemisinin (DHA) on TGF-β1-induced renal fibrosis in vitro and in vivo. Furthermore, m6A methyltransferase methyltransferase-like 3 (METTL3) was shown to be the main methyltransferase of m6A modification on MALAT1.
Collapse
Affiliation(s)
- Peihua Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Bo Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhi Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yao He
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yongchao Du
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yuhang Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Xiang Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| |
Collapse
|
42
|
Li Y, Ma Q, Li P, Wang J, Wang M, Fan Y, Wang T, Wang C, Wang T, Zhao B. Proteomics reveals different pathological processes of adipose tissue, liver, and skeletal muscle under insulin resistance. J Cell Physiol 2020; 235:6441-6461. [PMID: 32115712 DOI: 10.1002/jcp.29658] [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] [Received: 07/29/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes mellitus is the most common type of diabetes, and insulin resistance (IR) is its core pathological mechanism. Proteomics is an ingenious and promising Omics technology that can comprehensively describe the global protein expression profiling of body or specific tissue, and is widely applied to the study of molecular mechanisms of diseases. In this paper, we focused on insulin target organs: adipose tissue, liver, and skeletal muscle, and analyzed the different pathological processes of IR in these three tissues based on proteomics research. By literature studies, we proposed that the main pathological processes of IR among target organs were diverse, which showed unique characteristics and focuses. We further summarized the differential proteins in target organs which were verified to be related to IR, and discussed the proteins that may play key roles in the emphasized pathological processes, aiming at discovering potentially specific differential proteins of IR, and providing new ideas for pathological mechanism research of IR.
Collapse
Affiliation(s)
- Yaqi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Quantao Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingkang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Min Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
43
|
Hashemi Gheinani A, Bigger-Allen A, Wacker A, Adam RM. Systems analysis of benign bladder disorders: insights from omics analysis. Am J Physiol Renal Physiol 2020; 318:F901-F910. [PMID: 32116016 DOI: 10.1152/ajprenal.00496.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The signaling pathways and effectors that drive the response of the bladder to nonmalignant insults or injury are incompletely defined. Interrogation of biological systems has been revolutionized by the ability to generate high-content data sets that capture information on a variety of biomolecules in cells and tissues, from DNA to RNA to proteins. In oncology, such an approach has led to the identification of cancer subtypes, improved prognostic capability, and has provided a basis for precision treatment of patients. In contrast, systematic molecular characterization of benign bladder disorders has lagged behind, such that our ability to uncover novel therapeutic interventions or increase our mechanistic understanding of such conditions is limited. Here, we discuss existing literature on the application of omics approaches, including transcriptomics and proteomics, to urinary tract conditions characterized by pathological tissue remodeling. We discuss molecular pathways implicated in remodeling, challenges in the field, and aspirations for omics-based research in the future.
Collapse
Affiliation(s)
- Ali Hashemi Gheinani
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Alexander Bigger-Allen
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts.,Biological and Biomedical Sciences PhD Program, Harvard Medical School, Boston, Massachusetts
| | - Amanda Wacker
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts.,Florida State University, Tallahassee, Florida
| | - Rosalyn M Adam
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts.,Department of Surgery, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
44
|
Zhou LJ, Yang DW, Ou LN, Guo XR, Wu BL. Circulating Expression Level of LncRNA Malat1 in Diabetic Kidney Disease Patients and Its Clinical Significance. J Diabetes Res 2020; 2020:4729019. [PMID: 32832561 PMCID: PMC7421584 DOI: 10.1155/2020/4729019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/03/2020] [Accepted: 06/27/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Long noncoding RNA MALAT1 is closely related to diabetes and kidney diseases and is expected to be a new target for the diagnosis and treatment of diabetic nephropathy. OBJECTIVE This study aimed to explore the circulating expression level and significance of lncRNA Malat1 in patients with type 2 diabetes mellitus (T2DM) and diabetic kidney disease (DKD). METHODS Quantitative real-time PCR (qPCR) was conducted to assess the expression of lncRNA Malat1 in 20 T2DM patients, 27 DKD patients, and 14 healthy controls, and then, the clinical significance was analyzed. RESULTS LncRNA MALAT1 expression in peripheral blood mononuclear cells (PBMC) was significantly upregulated in T2DM and DKD groups when compared to control. Pearson's correlation analysis showed correlation of lncRNA MALAT1 levels with ACR, urine β2-microglobulin (β2-MG), urine α1-microglobulin (α1-MG), creatinine (Cr), and glycosylated hemoglobin (HbA1c), while negative with superoxide dismutase (SOD) (r = -0.388, P < 0.05). Binary regression analysis showed that ACR, creatinine, α1-MG, and LncRNA Malat1 were the risk factors for diabetic nephropathy with OR value of 1.166, 1.031, 1.031, and 2.019 (P < 0.05). The area under ROC curve (AUC) of DKD identified by the above indicators was 0.914, 0.643, 0.807, and 0.797, respectively. The AUC of Joint prediction probability of DKD recognition was 0.914, and the sensitivity and specificity of DKD diagnosis were 1.0 and 0.806, respectively. (Take ≥0.251 as the diagnostic cutoff point). CONCLUSION LncRNA Malat1 is highly expressed in DKD patients, and the combined detection of ACR, creatinine, α1-MG, and LncRNA Malat1 with diabetes mellitus may be the best way to diagnose diabetic nephropathy.
Collapse
Affiliation(s)
| | - Da-wei Yang
- Department of Endocrinology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China 533000
| | - Li-Na Ou
- Department of Endocrinology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China 533000
| | - Xing-Rong Guo
- Department of Endocrinology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China 533000
| | - Biao-liang Wu
- Department of Endocrinology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China 533000
| |
Collapse
|
45
|
Zhu B, Cheng X, Jiang Y, Cheng M, Chen L, Bao J, Tang X. Silencing of KCNQ1OT1 Decreases Oxidative Stress and Pyroptosis of Renal Tubular Epithelial Cells. Diabetes Metab Syndr Obes 2020; 13:365-375. [PMID: 32104033 PMCID: PMC7025682 DOI: 10.2147/dmso.s225791] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) can regulate the progression of DN. This research aimed to study the effect of lncRNA KCNQ1OT1 on the oxidative stress and pyroptosis of the renal tubular epithelial cells induced by high glucose (HG). METHODS RT-qPCR analysis detected the KCNQ1OT1 expression in serum with DN and HG-induced HK-2 cells, detect the expression of NLRP3, cleaved-caspase1, P-caspase1, IL-1β, p-IL-1β and GSDMD-N in HG-induced HK-2 cells, and confirm the transfection effects. The expression of NLRP3, cleaved-caspase1, P-caspase1, IL-1β, p-IL-1β and GSDMD-N in HG-induced HK-2 cells was also analyzed by Western blot analysis. ELISA assay detected the levels of TNF-α, IL-6 and MCP-1. The levels of ROS, MDA and SOD were determined by respective ELISA kits and ROS was also detected by the ROS assay kit (containing DCFH-DA). RESULTS We found that KCNQ1OT1 was increased in the plasma of patients with DN and HG-induced HK-2 cells and KCNQ1OT1 interference could decrease the inflammation, oxidative stress and pyroptosis of HG-induced HK-2 cells. In addition, KCNQ1OT1 directly targets miR-506-3p. MiR-506-3p was downregulated in the plasma of patients with DN and HG-induced HK-2 cells and KCNQ1OT1 interference promoted the expression of miR-506-3p. MiR-506-3p overexpression suppressed the inflammation, oxidative stress and pyroptosis of HG-induced HK-2 cells. CONCLUSION This study demonstrated that downregulation of KCNQ1OT1 inhibited the inflammation, oxidative stress and pyroptosis of HG-induced HK-2 cells by up-regulating the expression of miR-506-3p, which provide new insights into the treatment of DN.
Collapse
Affiliation(s)
- Bei Zhu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People’s Republic of China, Department of Endocrinology, Rugao People’s Hospital, Nantong226500, People’s Republic of China
| | - Xingbo Cheng
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou215006, People’s Republic of China
- Correspondence: Xingbo Cheng Department of Endocrinology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou215006, People’s Republic of China Email
| | - Yilan Jiang
- Department of Endocrinology, Rugao People’s Hospital, Nantong226500, People’s Republic of China
| | - Ming Cheng
- School of Rail Transportation, Soochow University, Suzhou215131, People’s Republic of China
- Ming Cheng School of Rail Transportation, Soochow University, 8 Ji Xue Road., Xiangcheng District, Suzhou215131, People’s Republic of China Email
| | - Luping Chen
- Department of Endocrinology, Rugao People’s Hospital, Nantong226500, People’s Republic of China
| | - Jiajun Bao
- Department of Endocrinology, Rugao People’s Hospital, Nantong226500, People’s Republic of China
| | - Xiaofeng Tang
- Department of Endocrinology, Rugao People’s Hospital, Nantong226500, People’s Republic of China
| |
Collapse
|
46
|
Characterization of Matricellular Protein Expression Signatures in Mechanistically Diverse Mouse Models of Kidney Injury. Sci Rep 2019; 9:16736. [PMID: 31723159 PMCID: PMC6854083 DOI: 10.1038/s41598-019-52961-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022] Open
Abstract
Fibrosis is the most common pathophysiological manifestation of Chronic Kidney Disease (CKD). It is defined as excessive deposition of extracellular matrix (ECM) proteins. Embedded within the ECM are a family of proteins called Matricellular Proteins (MCPs), which are typically expressed during chronic pathologies for ECM processing. As such, identifying potential MCPs in the pathological secretome of a damaged kidney could serve as diagnostic/therapeutic targets of fibrosis. Using published RNA-Seq data from two kidney injury mouse models of different etiologies, Folic Acid (FA) and Unilateral Ureteral Obstruction (UUO), we compared and contrasted the expression profile of various members from well-known MCP families during the Acute and Fibrotic injury phases. As a result, we identified common and distinct MCP expression signatures between both injury models. Bioinformatic analysis of their differentially expressed MCP genes revealed similar top annotation clusters from Molecular Function and Biological Process networks, which are those commonly involved in fibrosis. Using kidney lysates from FA- and UUO-injured mice, we selected MCP genes from our candidate list to confirm mRNA expression by Western Blot, which correlated with injury progression. Understanding the expressions of MCPs will provide important insight into the processes of kidney repair, and may validate MCPs as biomarkers and/or therapeutic targets of CKD.
Collapse
|
47
|
Wu H, Lai CF, Chang-Panesso M, Humphreys BD. Proximal Tubule Translational Profiling during Kidney Fibrosis Reveals Proinflammatory and Long Noncoding RNA Expression Patterns with Sexual Dimorphism. J Am Soc Nephrol 2019; 31:23-38. [PMID: 31537650 DOI: 10.1681/asn.2019040337] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/01/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Proximal tubule injury can initiate CKD, with progression rates that are approximately 50% faster in males versus females. The precise transcriptional changes in this nephron segment during fibrosis and potential differences between sexes remain undefined. METHODS We generated mice with proximal tubule-specific expression of an L10a ribosomal subunit protein fused with enhanced green fluorescent protein. We performed unilateral ureteral obstruction surgery on four male and three female mice to induce inflammation and fibrosis, collected proximal tubule-specific and bulk cortex mRNA at day 5 or 10, and sequenced samples to a depth of 30 million reads. We applied computational methods to identify sex-biased and shared molecular responses to fibrotic injury, including up- and downregulated long noncoding RNAs (lncRNAs) and transcriptional regulators, and used in situ hybridization to validate critical genes and pathways. RESULTS We identified >17,000 genes in each proximal tubule group, including 145 G-protein-coupled receptors. More than 700 transcripts were differentially expressed in the proximal tubule of males versus females. The >4000 genes displaying altered expression during fibrosis were enriched for proinflammatory and profibrotic pathways. Our identification of nearly 150 differentially expressed proximal tubule lncRNAs during fibrosis suggests they may have unanticipated regulatory roles. Network analysis prioritized proinflammatory and profibrotic transcription factors such as Irf1, Nfkb1, and Stat3 as drivers of fibrosis progression. CONCLUSIONS This comprehensive transcriptomic map of the proximal tubule revealed sexually dimorphic gene expression that may reflect sex-related disparities in CKD, proinflammatory gene modules, and previously unappreciated proximal tubule-specific bidirectional lncRNA regulation.
Collapse
Affiliation(s)
- Haojia Wu
- Division of Nephrology.,Departments of Medicine and
| | - Chun-Fu Lai
- Division of Nephrology.,Departments of Medicine and.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipai, Taiwan
| | | | - Benjamin D Humphreys
- Division of Nephrology, .,Departments of Medicine and.,Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri; and
| |
Collapse
|
48
|
Hewitson TD, Holt SG, Samuel CS, Wigg B, Smith ER. Profiling histone modifications in the normal mouse kidney and after unilateral ureteric obstruction. Am J Physiol Renal Physiol 2019; 317:F606-F615. [DOI: 10.1152/ajprenal.00262.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Posttranslational modification of nucleosomal histones is a major determinant of chromatin structure and gene activity. In the present study, we hypothesized that unilateral ureteric obstruction (UUO), a widely used model of tubulointerstitial injury, would be associated with a distinct pattern of histone modifications (marks) in the kidney. Mass spectrometry was used to profile 63 different histone marks in normal mouse kidneys and those after 10 days of UUO. A subsequent histochemical analysis further examined examples of specific marks that changed significantly after UUO for which antisera are available. Histone marks were much more widely distributed and abundant in the normal kidney than is usually appreciated. Although aggregate analysis of the mass spectrometry results revealed net differences between control and UUO groups, residue-specific variations were subtle. Of the 16/63 significant changes ( P < 0.05), only 8 changes were quantitatively different by >5%. Nevertheless, we identified several that are not usually examined in the kidney, including marks in the globular domain of core histones (H3:K79), linker histones (H1.4), and histone variants (H3.1:K27 and H3.3:K27). In several cases, there were complementary changes in different marks on the same amino acid. Using H3:K79ME2 as an example, mark enrichment was heterogeneous but largely colocalized with active transcription in a subset of tubular pathology. In conclusion, our study highlights the importance of unbiased screening in examining histone marks. Simultaneous changes in multiple marks on the same amino acid indicate a coordinated histone mark signature. The heterogeneous enrichment of marks, even within the same tubule, highlights the importance of regulatory context.
Collapse
Affiliation(s)
- Timothy D. Hewitson
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Stephen G. Holt
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Chrishan S. Samuel
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Belinda Wigg
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Edward R. Smith
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
49
|
Reichelt-Wurm S, Wirtz T, Chittka D, Lindenmeyer M, Reichelt RM, Beck S, Politis P, Charonis A, Kretz M, Huber TB, Liu S, Banas B, Banas MC. Glomerular expression pattern of long non-coding RNAs in the type 2 diabetes mellitus BTBR mouse model. Sci Rep 2019; 9:9765. [PMID: 31278342 PMCID: PMC6611801 DOI: 10.1038/s41598-019-46180-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 06/11/2019] [Indexed: 11/09/2022] Open
Abstract
The prevalence of type 2 diabetes mellitus (T2DM) and by association diabetic nephropathy (DN) will continuously increase in the next decades. Nevertheless, the underlying molecular mechanisms are largely unknown and studies on the role of new actors like long non-coding RNAs (lncRNAs) barely exist. In the present study, the inherently insulin-resistant mouse strain "black and tan, brachyuric" (BTBR) served as T2DM model. While wild-type mice do not exhibit pathological changes, leptin-deficient diabetic animals develop a severe T2DM accompanied by a DN, which closely resembles the human phenotype. We analyzed the glomerular expression of lncRNAs from wild-type and diabetic BTBR mice (four, eight, 16, and 24 weeks) applying the "GeneChip Mouse Whole Transcriptome 1.0 ST" array. This microarray covered more lncRNA gene loci than any other array before. Over the observed time, our data revealed differential expression patterns of 1746 lncRNAs, which markedly differed from mRNAs. We identified protein-coding and non-coding genes, that were not only co-located but also co-expressed, indicating a potentially cis-acting function of these lncRNAs. In vitro-experiments strongly suggested a cell-specific expression of these lncRNA-mRNA-pairs. Additionally, protein-coding genes, being associated with significantly regulated lncRNAs, were enriched in various biological processes and pathways, that were strongly linked to diabetes.
Collapse
Affiliation(s)
| | - Tobias Wirtz
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Dominik Chittka
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Maja Lindenmeyer
- Nephrological Center, Medical Clinic and Policlinic IV, University Hospital of Munich, Munich, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robert M Reichelt
- Department of Biochemistry, Genetics and Microbiology, Institute of Microbiology, University of Regensburg, Regensburg, Germany
| | - Sebastian Beck
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Panagiotis Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Aristidis Charonis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Markus Kretz
- Institute of Biochemistry, Genetics and Microbiology, University of Regensburg, Regensburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Shuya Liu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Miriam C Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
50
|
Brandenburger T, Salgado Somoza A, Devaux Y, Lorenzen JM. Noncoding RNAs in acute kidney injury. Kidney Int 2019; 94:870-881. [PMID: 30348304 DOI: 10.1016/j.kint.2018.06.033] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/30/2018] [Accepted: 06/11/2018] [Indexed: 02/09/2023]
Abstract
Acute kidney injury (AKI) is an important health issue concerning ∼50% of patients treated in intensive care units. AKI mainly occurs after sepsis, acute ischemia, nephrotoxicity, or hypoxia and leads to severe damage of the kidney and to an increased risk of mortality. The diagnosis of AKI is currently based on creatinine urea levels and diuresis. Yet, novel markers may improve the accuracy of this diagnosis at an early stage of the disease, thereby allowing early prevention and therapy, ultimately leading to a reduction in the need for renal replacement therapy and decreased mortality. Non-protein-coding RNAs or noncoding RNAs are central players in development and disease. They are important regulatory molecules that allow a fine-tuning of gene expression and protein synthesis. This regulation is necessary to maintain homeostasis, and its dysregulation is often associated with disease development. Noncoding RNAs are present in the kidney and in body fluids and their expression is modulated during AKI. This review article assembles the current knowledge of the role of noncoding RNAs, including microRNAs, long noncoding RNAs and circular RNAs, in the pathogenesis of AKI. Their potential as biomarkers and therapeutic targets as well as the challenges to translate research findings to clinical application are discussed. Although microRNAs have entered clinical testing, preclinical and clinical trials are needed before long noncoding RNAs and circular RNAs may be considered as useful biomarkers or therapeutic targets of AKI.
Collapse
Affiliation(s)
- Timo Brandenburger
- Department of Anesthesiology, University Hospital Duesseldorf, Duesseldorf, Germany.
| | - Antonio Salgado Somoza
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Johan M Lorenzen
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|