1
|
Kan S, Hou Q, Shi J, Zhang M, Xu F, Liu Z, Jiang S. EHHADH deficiency regulates pexophagy and accelerates tubulointerstitial injury in diabetic kidney disease. Cell Death Discov 2024; 10:289. [PMID: 38879653 PMCID: PMC11180138 DOI: 10.1038/s41420-024-02066-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/19/2024] Open
Abstract
Peroxisomal L-bifunctional enzyme (EHHADH) plays a role in the classic peroxisomal fatty acid β-oxidation pathway; however, the relationship between EHHADH expression and diabetic kidney disease has not been well understood. Here, we found that endogenous EHHADH levels were strongly correlated with the progression and severity of diabetic nephropathy in T2D patients. EHHADH knockout mice exhibited worsened renal tubular injury in diabetic mice. Furthermore, EHHADH is a modulator of pexophagy. In renal tubular epithelial cells (RTECs) in vitro, the knockdown of EHHADH induced a dramatic loss of peroxisomes. The loss of peroxisomes in EHHADH-deficient RTECs was restored by either an autophagic inhibitor 3-methyladenine or bafilomycin A1 both in vitro and in vivo. NBR1 was required for pexophagy in EHHADH-knockdown cells, where the level of reactive oxygen species (ROS) was increased, while inhibition of ROS blocked pexophagy. In summary, our findings revealed EHHADH deficiency accelerated renal injury in DKD as a modulator of pexophagy.
Collapse
Affiliation(s)
- Shuyan Kan
- National Clinical Research Center for Kidney Disease, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qing Hou
- National Clinical Research Center for Kidney Disease, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jinsong Shi
- National Clinical Research Center for Kidney Disease, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mingchao Zhang
- National Clinical Research Center for Kidney Disease, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Feng Xu
- National Clinical Research Center for Kidney Disease, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhihong Liu
- National Clinical Research Center for Kidney Disease, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Song Jiang
- National Clinical Research Center for Kidney Disease, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Duan X, Chen C, Liu X, Wang T, Feng S, Li J, Li G. Interference of periostin attenuates pathological changes, proinflammatory markers and renal fibrosis in diabetic kidney injury. Genes Genomics 2023; 45:1389-1397. [PMID: 37248423 DOI: 10.1007/s13258-023-01400-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/11/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a prevalent complication of diabetes, in which inflammation and fibrosis are the significant pathogenesis. Periostin is a matricellular protein that functions on stabilizing the extracellular matrix by binding to integrins during development. This study aimed to explored the role of periostin in DN. METHODS The animal and cell models of DN were constructed in streptozocin (STZ)-induced mice and high glucose-challenged human mesangial cells (HMCs). The role of periostin in pathological changes, inflammation and fibrosis in DN was investigated through biochemical detection, HE and Masson staining and scores, western blot, enzyme‑linked immunosorbent assay (ELISA) and real-time quantitative PCR (RT-qPCR) assays. RESULTS Knockdown of periostin counteracted the STZ-induced the ratio of kidney weight and body weight, and the concentrations of urine albumin excretion (UAE), serum creatinine (Scr), urine albumin/creatinine ratio (UACR) and blood urea nitrogen (BUN) in mice. Moreover, silencing of periostin alleviated the pathological manifestations and reduced the concentrations of IL-6, TNF-α and IL-1β in mice kidney tissues and sera. Also, downregulation of periostin decreased the relative protein expression of fibronectin, collagen IV and α-SMA in kidney tissues. Meanwhile, interference of periostin attenuated the levels of pro-inflammation factors and the expressions of fibrosis markers in HG-induced HMCs. CONCLUSION Interference of periostin resisted DN via attenuating the pro-inflammatory cytokines release and renal fibrosis in diabetic kidney injury. Our study establishes a basis for its further study and underlying application in clinical practice in diagnosing and treating diabetic kidney injury or other relevant diseases.
Collapse
Affiliation(s)
- Xiaoting Duan
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Cheng Chen
- The Second Department of Oncology, Affiliated Hospital of Hebei Engineering University, Hebei, 056000, China
| | - Xiaoli Liu
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Taoxia Wang
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Shuning Feng
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Jianwei Li
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Guiying Li
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China.
| |
Collapse
|
3
|
Shelke V, Yelgonde V, Kale A, Lech M, Gaikwad AB. Epigenetic regulation of mitochondrial-endoplasmic reticulum dynamics in kidney diseases. J Cell Physiol 2023; 238:1716-1731. [PMID: 37357431 DOI: 10.1002/jcp.31058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 06/27/2023]
Abstract
Kidney diseases are serious health problems affecting >800 million individuals worldwide. The high number of affected individuals and the severe consequences of kidney dysfunction demand an intensified effort toward more effective prevention and treatment. The pathophysiology of kidney diseases is complex and comprises diverse organelle dysfunctions including mitochondria and endoplasmic reticulum (ER). The recent findings prove interactions between the ER membrane and nearly all cell compartments and give new insights into molecular events involved in cellular mechanisms in health and disease. Interactions between the ER and mitochondrial membranes, known as the mitochondria-ER contacts regulate kidney physiology by interacting with each other via membrane contact sites (MCS). ER controls mitochondrial dynamics through ER stress sensor proteins or by direct communication via mitochondria-associated ER membrane to activate signaling pathways such as apoptosis, calcium transport, and autophagy. More importantly, these organelle dynamics are found to be regulated by several epigenetic mechanisms such as DNA methylation, histone modifications, and noncoding RNAs and can be a potential therapeutic target against kidney diseases. However, a thorough understanding of the role of epigenetic regulation of organelle dynamics and their functions is not well understood. Therefore, this review will unveil the role of epigenetic mechanisms in regulating organelle dynamics during various types of kidney diseases. Moreover, we will also shed light on different stress origins in organelles leading to kidney disease. Henceforth, by understanding this we can target epigenetic mechanisms to maintain/control organelle dynamics and serve them as a novel therapeutic approach against kidney diseases.
Collapse
Affiliation(s)
- Vishwadeep Shelke
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Vinayak Yelgonde
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Ajinath Kale
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Maciej Lech
- Department of Internal Medicine IV, Division of Nephrology, Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, Rajasthan, India
| |
Collapse
|
4
|
Venkatesan M, Zhang N, Marteau B, Yajima Y, De Zarate Garcia NO, Fang Z, Hu T, Cai S, Ford A, Olszewski H, Borst A, Coskun AF. Spatial subcellular organelle networks in single cells. Sci Rep 2023; 13:5374. [PMID: 37005468 PMCID: PMC10067843 DOI: 10.1038/s41598-023-32474-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
Organelles play important roles in human health and disease, such as maintaining homeostasis, regulating growth and aging, and generating energy. Organelle diversity in cells not only exists between cell types but also between individual cells. Therefore, studying the distribution of organelles at the single-cell level is important to understand cellular function. Mesenchymal stem cells are multipotent cells that have been explored as a therapeutic method for treating a variety of diseases. Studying how organelles are structured in these cells can answer questions about their characteristics and potential. Herein, rapid multiplexed immunofluorescence (RapMIF) was performed to understand the spatial organization of 10 organelle proteins and the interactions between them in the bone marrow (BM) and umbilical cord (UC) mesenchymal stem cells (MSCs). Spatial correlations, colocalization, clustering, statistical tests, texture, and morphological analyses were conducted at the single cell level, shedding light onto the interrelations between the organelles and comparisons of the two MSC subtypes. Such analytics toolsets indicated that UC MSCs exhibited higher organelle expression and spatially spread distribution of mitochondria accompanied by several other organelles compared to BM MSCs. This data-driven single-cell approach provided by rapid subcellular proteomic imaging enables personalized stem cell therapeutics.
Collapse
Affiliation(s)
- Mythreye Venkatesan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nicholas Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Benoit Marteau
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yukina Yajima
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nerea Ortiz De Zarate Garcia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Departamento de Bioingenieria e Ingenieria Aeroespacial, Universidad Carlos III de Madrid, Getafe, Spain
| | - Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Thomas Hu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shuangyi Cai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Adam Ford
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Harrison Olszewski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrew Borst
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
5
|
Han YP, Liu LJ, Yan JL, Chen MY, Meng XF, Zhou XR, Qian LB. Autophagy and its therapeutic potential in diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1139444. [PMID: 37020591 PMCID: PMC10067862 DOI: 10.3389/fendo.2023.1139444] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
Diabetic nephropathy (DN), the leading cause of end-stage renal disease, is the most significant microvascular complication of diabetes and poses a severe public health concern due to a lack of effective clinical treatments. Autophagy is a lysosomal process that degrades damaged proteins and organelles to preserve cellular homeostasis. Emerging studies have shown that disorder in autophagy results in the accumulation of damaged proteins and organelles in diabetic renal cells and promotes the development of DN. Autophagy is regulated by nutrient-sensing pathways including AMPK, mTOR, and Sirt1, and several intracellular stress signaling pathways such as oxidative stress and endoplasmic reticulum stress. An abnormal nutritional status and excess cellular stresses caused by diabetes-related metabolic disorders disturb the autophagic flux, leading to cellular dysfunction and DN. Here, we summarized the role of autophagy in DN focusing on signaling pathways to modulate autophagy and therapeutic interferences of autophagy in DN.
Collapse
Affiliation(s)
- Yu-Peng Han
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Li-Juan Liu
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Jia-Lin Yan
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Meng-Yuan Chen
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xiang-Fei Meng
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xin-Ru Zhou
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Ling-Bo Qian
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
6
|
Chen Q, Hong Z, Chen Z, Chen Y, Liu D. CircRNA expression profiles and functional analysis in a mouse model of chronic intermittent hypoxia-induced renal injury: new insight into pathogenesis. PeerJ 2023; 11:e14957. [PMID: 36874972 PMCID: PMC9983420 DOI: 10.7717/peerj.14957] [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: 11/14/2022] [Accepted: 02/05/2023] [Indexed: 03/04/2023] Open
Abstract
Increasing evidence has demonstrated that circular RNAs (circRNAs) play crucial roles in the pathogenesis of multiple diseases. However, the functions of circRNAs in renal injury induced by obstructive sleep apnea (OSA) are poorly understood. The aim of this current study is to identify the global changes of circRNAs expression in OSA-induced renal damage. The mouse model of OSA treated by chronic intermittent hypoxia (CIH) was established. We assessed the expression profiles of circRNAs in CIH caused renal injury by microarray analysis. Bioinformatic analyses were further performed by us to assess those differentially expressed circRNAs. Quantitative realtime PCR (qRT-PCR) were then conducted to assure the data of microarray. Finally, a circRNA-miRNA -mRNA competing endogenous RNA (ceRNA) regulatory network was constructed. We found 11 upregulated and 13 downregulated circRNAs in CIH-induced renal injury. The qRT-PCR validated that the six selected circRNAs were identical to the results of microarray. Both Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were further employed to annotate the potential functions of dysregulated circRNAs. Finally, we established a ceRNA network to predict the target genes of circRNAs. In general, our results first illustrate that circRNAs are aberrantly expressed in OSA-induced renal injury, which might aid in offering novel genetic insights into this disease and potential therapeutic targets for OSA-associated chronic kidney disease.
Collapse
Affiliation(s)
- Qingshi Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhenzhen Hong
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhiyu Chen
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yanfeng Chen
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Dexin Liu
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
7
|
Tsai JL, Chen CH, Wu MJ, Tsai SF. New Approaches to Diabetic Nephropathy from Bed to Bench. Biomedicines 2022; 10:biomedicines10040876. [PMID: 35453626 PMCID: PMC9031931 DOI: 10.3390/biomedicines10040876] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 02/01/2023] Open
Abstract
Diabetic nephropathy (DN) is the main cause of end-stage kidney disease (ESKD). DN-related ESKD has the worst prognosis for survival compared with other causes. Due to the complex mechanisms of DN and the heterogeneous presentations, unmet needs exist for the renal outcome of diabetes mellitus. Clinical evidence for treating DN is rather solid. For example, the first Kidney Disease: Improving Global Outcomes (KDIGO) guideline was published in October 2020: KDIGO Clinical Practice Guideline for Diabetes Management in Chronic Kidney Disease. In December of 2020, the International Society of Nephrology published 60 (+1) breakthrough discoveries in nephrology. Among these breakthroughs, four important ones after 1980 were recognized, including glomerular hyperfiltration theory, renal protection by renin-angiotensin system inhibition, hypoxia-inducible factor, and sodium-glucose cotransporter 2 inhibitors. Here, we present a review on the pivotal and new mechanisms of DN from the implications of clinical studies and medications.
Collapse
Affiliation(s)
- Jun-Li Tsai
- Division of Family Medicine, Cheng Ching General Hospital, Taichung 407, Taiwan;
- Division of Family Medicine, Cheng Ching Rehabilitation Hospital, Taichung 407, Taiwan
| | - Cheng-Hsu Chen
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan; (C.-H.C.); (M.-J.W.)
- Department of Life Science, Tunghai University, Taichung 407, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan; (C.-H.C.); (M.-J.W.)
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Shang-Feng Tsai
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan; (C.-H.C.); (M.-J.W.)
- Department of Life Science, Tunghai University, Taichung 407, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Correspondence:
| |
Collapse
|
8
|
Molitoris BA, Sandoval RM, Yadav SPS, Wagner MC. Albumin Uptake and Processing by the Proximal Tubule: Physiologic, Pathologic and Therapeutic Implications. Physiol Rev 2022; 102:1625-1667. [PMID: 35378997 PMCID: PMC9255719 DOI: 10.1152/physrev.00014.2021] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
For nearly 50 years the proximal tubule (PT) has been known to reabsorb, process, and either catabolize or transcytose albumin from the glomerular filtrate. Innovative techniques and approaches have provided insights into these processes. Several genetic diseases, nonselective PT cell defects, chronic kidney disease (CKD), and acute PT injury lead to significant albuminuria, reaching nephrotic range. Albumin is also known to stimulate PT injury cascades. Thus, the mechanisms of albumin reabsorption, catabolism, and transcytosis are being reexamined with the use of techniques that allow for novel molecular and cellular discoveries. Megalin, a scavenger receptor, cubilin, amnionless, and Dab2 form a nonselective multireceptor complex that mediates albumin binding and uptake and directs proteins for lysosomal degradation after endocytosis. Albumin transcytosis is mediated by a pH-dependent binding affinity to the neonatal Fc receptor (FcRn) in the endosomal compartments. This reclamation pathway rescues albumin from urinary losses and cellular catabolism, extending its serum half-life. Albumin that has been altered by oxidation, glycation, or carbamylation or because of other bound ligands that do not bind to FcRn traffics to the lysosome. This molecular sorting mechanism reclaims physiological albumin and eliminates potentially toxic albumin. The clinical importance of PT albumin metabolism has also increased as albumin is now being used to bind therapeutic agents to extend their half-life and minimize filtration and kidney injury. The purpose of this review is to update and integrate evolving information regarding the reabsorption and processing of albumin by proximal tubule cells including discussion of genetic disorders and therapeutic considerations.
Collapse
Affiliation(s)
- Bruce A. Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Dept.of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Ruben M. Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Shiv Pratap S. Yadav
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Mark C. Wagner
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
9
|
Wang D, Kang L, Chen C, Guo J, Du L, Zhou D, Li G, Zhang Y, Mi X, Zhang M, Liu S, Tan X. Loss of legumain induces premature senescence and mediates aging-related renal fibrosis. Aging Cell 2022; 21:e13574. [PMID: 35195326 PMCID: PMC8920435 DOI: 10.1111/acel.13574] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 12/14/2022] Open
Abstract
Aging is an independent risk factor for acute kidney injury and subsequent chronic kidney diseases, while the underlying mechanism is still elusive. Here, we found that renal tubules highly express a conserved lysosomal endopeptidase, legumain, which is significantly downregulated with the growing of age. Tubule‐specific legumain‐knockout mice exhibit spontaneous renal interstitial fibrosis from the 3rd month. In the tubule‐specific legumain‐knockout mice and the cultured legumain‐knockdown HK‐2 cells, legumain deficiency induces the activation of tubular senescence and thus increases the secretion of profibrotic senescence‐associated cytokines, which in turn accelerates the activation of fibroblasts. Blockage of senescence mitigates the fibrotic lesion caused by legumain deficiency. Mechanistically, we found that silencing down of legumain leads to the elevated lysosome pH value, enlargement of lysosome size, and increase of lysosomal voltage dependent membrane channel proteins. Either legumain downregulation or aging alone induces the activation of nuclear transcription factors EB (TFEB) while it fails to further upregulate in the elderly legumain‐knockdown tubules, accompanied with impaired mitophagy and increased mitochondrial ROS (mtROS) accumulation. Therapeutically, supplementation of exosomal legumain ameliorated fibronectin and collagen I production in an in vitro coculture system of tubular cells and fibroblasts. Altogether, our data demonstrate that loss of legumain in combined with aging dysregulates lysosomal homeostasis, although either aging or legumain deficiency alone induces lysosome adaptation via stimulating lysosomal biogenesis. Consequently, impaired mitophagy leads to mtROS accumulation and therefore activates tubular senescence and boosts the interstitial fibrosis.
Collapse
Affiliation(s)
- Dekun Wang
- Department of Pathology School of Medicine Nankai University Tianjin China
| | - Lichun Kang
- Department of Pathology School of Medicine Nankai University Tianjin China
| | - Chuan'ai Chen
- Department of Pathology School of Medicine Nankai University Tianjin China
| | - Jiasen Guo
- College of Life Science Nankai University Tianjin China
| | - Lingfang Du
- Department of Pathology School of Medicine Nankai University Tianjin China
| | - Donghui Zhou
- Department of Pathology School of Medicine Nankai University Tianjin China
| | - Gang Li
- Nephrology Division The Second Hospital of Tianjin Medical University Tianjin China
| | - Yuying Zhang
- Department of Pathology School of Medicine Nankai University Tianjin China
| | - Xue Mi
- Department of Pathology School of Medicine Nankai University Tianjin China
| | - Mianzhi Zhang
- Dongfang Hospital of Beijing University of Chinese medicine Beijing China
| | - Shuxia Liu
- Hebei Key Laboratory of Nephrology Department of Pathology Hebei Medical University Shijiazhuang China
| | - Xiaoyue Tan
- Department of Pathology School of Medicine Nankai University Tianjin China
| |
Collapse
|
10
|
Yao T, Su W, Han S, Lu Y, Xu Y, Chen M, Wang Y. Recent Advances in Traditional Chinese Medicine for Treatment of Podocyte Injury. Front Pharmacol 2022; 13:816025. [PMID: 35281899 PMCID: PMC8914202 DOI: 10.3389/fphar.2022.816025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/12/2022] [Indexed: 12/03/2022] Open
Abstract
Podocyte is also called glomerular epithelial cell, which has been considered as the final gatekeeper of glomerular filtration barrier (GFB). As a major contributor to proteinuria, podocyte injury underlies a variety of glomerular diseases and becomes the challenge to patients and their families in general. At present, the therapeutic methods of podocyte injury mainly include angiotensin-converting enzyme inhibitors or angiotensin receptor blockers, steroid and immunosuppressive medications. Nevertheless, the higher cost and side effects seriously disturb patients with podocyte injury. Promisingly, traditional Chinese medicine (TCM) has received an increasing amount of attention from different countries in the treatment of podocyte injury by invigorating spleen and kidney, clearing heat and eliminating dampness, as well enriching qi and activating blood. Therefore, we searched articles published in peer-reviewed English-language journals through Google Scholar, PubMed, Web of Science, and Science Direct. The protective effects of active ingredients, herbs, compound prescriptions, acupuncture and moxibustion for treatment of podocyte injury were further summarized and analyzed. Meanwhile, we discussed feasible directions for future development, and analyzed existing deficiencies and shortcomings of TCM in the treatment of podocyte injury. In conclusion, this paper shows that TCM treatments can serve as promising auxiliary therapeutic methods for the treatment of podocyte injury.
Collapse
Affiliation(s)
- Tianwen Yao
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenxiang Su
- Department of Nephrology, The People’s Hospital of Mengzi, Mengzi, China
| | - Shisheng Han
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Lu
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqiu Xu
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Chen
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Wang
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yi Wang,
| |
Collapse
|
11
|
Tserga A, Pouloudi D, Saulnier-Blache JS, Stroggilos R, Theochari I, Gakiopoulou H, Mischak H, Zoidakis J, Schanstra JP, Vlahou A, Makridakis M. Proteomic Analysis of Mouse Kidney Tissue Associates Peroxisomal Dysfunction with Early Diabetic Kidney Disease. Biomedicines 2022; 10:biomedicines10020216. [PMID: 35203426 PMCID: PMC8869654 DOI: 10.3390/biomedicines10020216] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
Background: The absence of efficient inhibitors for diabetic kidney disease (DKD) progression reflects the gaps in our understanding of DKD molecular pathogenesis. Methods: A comprehensive proteomic analysis was performed on the glomeruli and kidney cortex of diabetic mice with the subsequent validation of findings in human biopsies and omics datasets, aiming to better understand the underlying molecular biology of early DKD development and progression. Results: LC–MS/MS was employed to analyze the kidney proteome of 2 DKD models: Ins2Akita (early and late DKD) and db/db mice (late DKD). The abundance of detected proteins was defined. Pathway analysis of differentially expressed proteins in the early and late DKD versus the respective controls predicted dysregulation in DKD hallmarks (peroxisomal lipid metabolism and β-oxidation), supporting the functional relevance of the findings. Comparing the observed protein changes in early and late DKD, the consistent upregulation of 21 and downregulation of 18 proteins was detected. Among these were downregulated peroxisomal and upregulated mitochondrial proteins. Tissue sections from 16 DKD patients were analyzed by IHC confirming our results. Conclusion: Our study shows an extensive differential expression of peroxisomal proteins in the early stages of DKD that persists regardless of the disease severity, providing new perspectives and potential markers of diabetic kidney dysfunction.
Collapse
Affiliation(s)
- Aggeliki Tserga
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (R.S.); (J.Z.)
| | - Despoina Pouloudi
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.P.); (I.T.); (H.G.)
| | - Jean Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France;
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Rafael Stroggilos
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (R.S.); (J.Z.)
| | - Irene Theochari
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.P.); (I.T.); (H.G.)
| | - Harikleia Gakiopoulou
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.P.); (I.T.); (H.G.)
| | | | - Jerome Zoidakis
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (R.S.); (J.Z.)
| | - Joost Peter Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France;
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
- Correspondence: (J.P.S.); (A.V.); (M.M.); Tel.: +33-5-31224078 (J.P.S.); +30-210-6597506 (A.V.); +30-210-6597485 (M.M.)
| | - Antonia Vlahou
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (R.S.); (J.Z.)
- Correspondence: (J.P.S.); (A.V.); (M.M.); Tel.: +33-5-31224078 (J.P.S.); +30-210-6597506 (A.V.); +30-210-6597485 (M.M.)
| | - Manousos Makridakis
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (R.S.); (J.Z.)
- Correspondence: (J.P.S.); (A.V.); (M.M.); Tel.: +33-5-31224078 (J.P.S.); +30-210-6597506 (A.V.); +30-210-6597485 (M.M.)
| |
Collapse
|
12
|
Ungur RA, Borda IM, Codea RA, Ciortea VM, Năsui BA, Muste S, Sarpataky O, Filip M, Irsay L, Crăciun EC, Căinap S, Jivănescu DB, Pop AL, Singurean VE, Crișan M, Groza OB, Martiș (Petruț) GS. A Flavonoid-Rich Extract of Sambucus nigra L. Reduced Lipid Peroxidation in a Rat Experimental Model of Gentamicin Nephrotoxicity. MATERIALS (BASEL, SWITZERLAND) 2022; 15:772. [PMID: 35160718 PMCID: PMC8837157 DOI: 10.3390/ma15030772] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 12/04/2022]
Abstract
The use of gentamicin (GM) is limited due to its nephrotoxicity mediated by oxidative stress. This study aimed to evaluate the capacity of a flavonoid-rich extract of Sambucus nigra L. elderflower (SN) to inhibit lipoperoxidation in GM-induced nephrotoxicity. The HPLC analysis of the SN extract recorded high contents of rutin (463.2 ± 0.0 mg mL-1), epicatechin (9.0 ± 1.1 µg mL-1), and ferulic (1.5 ± 0.3 µg mL-1) and caffeic acid (3.6 ± 0.1 µg mL-1). Thirty-two Wistar male rats were randomized into four groups: a control group (C) (no treatment), GM group (100 mg kg-1 bw day-1 GM), GM+SN group (100 mg kg-1 bw day-1 GM and 1 mL SN extract day-1), and SN group (1 mL SN extract day-1). Lipid peroxidation, evaluated by malondialdehyde (MDA), and antioxidant enzymes activity-superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX)-were recorded in renal tissue after ten days of experimental treatment. The MDA level was significantly higher in the GM group compared to the control group (p < 0.0001), and was significantly reduced by SN in the GM+SN group compared to the GM group (p = 0.021). SN extract failed to improve SOD, CAT, and GPX activity in the GM+SN group compared to the GM group (p > 0.05), and its action was most probably due to the ability of flavonoids (rutin, epicatechin) and ferulic and caffeic acids to inhibit synthesis and neutralize reactive species, to reduce the redox-active iron pool, and to inhibit lipid peroxidation. In this study, we propose an innovative method for counteracting GM nephrotoxicity with a high efficiency and low cost, but with the disadvantage of the multifactorial environmental variability of the content of SN extracts.
Collapse
Affiliation(s)
- Rodica Ana Ungur
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Ileana Monica Borda
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Răzvan Andrei Codea
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania;
| | - Viorela Mihaela Ciortea
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Bogdana Adriana Năsui
- Department of Community Health, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Sevastița Muste
- Food Engineering Department, University of Agricultural Sciences and Veterinary Medicine, 64 Calea Floresti, 400509 Cluj-Napoca, Romania; (S.M.); (G.S.M.)
| | - Orsolya Sarpataky
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania;
| | - Miuța Filip
- Raluca Ripan Institute for Research in Chemistry, Babeş-Bolyai University, 30 Fântânele Street, 400294 Cluj-Napoca, Romania;
| | - Laszlo Irsay
- Department of Medical Specialties, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (R.A.U.); (V.M.C.); (L.I.)
| | - Elena Cristina Crăciun
- Department of Pharmaceutical Biochemistry and Clinical Laboratory, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur Street, 400349 Cluj-Napoca, Romania;
| | - Simona Căinap
- Department of Mother and Child, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Delia Bunea Jivănescu
- Department of Internal Medicine, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania;
| | - Anca Lucia Pop
- Department of Clinical Laboratory, Food Safety, Nutrition, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020945 Bucharest, Romania;
| | - Victoria Emilia Singurean
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Maria Crișan
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Oana Bianca Groza
- Department of Morphological Sciences, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania; (V.E.S.); (M.C.); (O.B.G.)
| | - Georgiana Smaranda Martiș (Petruț)
- Food Engineering Department, University of Agricultural Sciences and Veterinary Medicine, 64 Calea Floresti, 400509 Cluj-Napoca, Romania; (S.M.); (G.S.M.)
| |
Collapse
|
13
|
Yadav SPS, Sandoval RM, Zhao J, Huang Y, Wang E, Kumar S, Campos-Bilderback SB, Rhodes G, Mechref Y, Molitoris BA, Wagner MC. Mechanism of how carbamylation reduces albumin binding to FcRn contributing to increased vascular clearance. Am J Physiol Renal Physiol 2021; 320:F114-F129. [PMID: 33283642 PMCID: PMC7847050 DOI: 10.1152/ajprenal.00428.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/12/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease results in high serum urea concentrations leading to excessive protein carbamylation, primarily albumin. This is associated with increased cardiovascular disease and mortality. Multiple methods were used to address whether carbamylation alters albumin metabolism. Intravital two-photon imaging of the Munich Wistar Frömter (MWF) rat kidney and liver allowed us to characterize filtration and proximal tubule uptake and liver uptake. Microscale thermophoresis enabled quantification of cubilin (CUB7,8 domain) and FcRn binding. Finally, multiple biophysical methods including dynamic light scattering, small-angle X-ray scattering, LC-MS/MS and in silico analyses were used to identify the critical structural alterations and amino acid modifications of rat albumin. Carbamylation of albumin reduced binding to CUB7,8 and FcRn in a dose-dependent fashion. Carbamylation markedly increased vascular clearance of carbamylated rat serum albumin (cRSA) and altered distribution of cRSA in both the kidney and liver at 16 h post intravenous injection. By evaluating the time course of carbamylation and associated charge, size, shape, and binding parameters in combination with in silico analysis and mass spectrometry, the critical binding interaction impacting carbamylated albumin's reduced FcRn binding was identified as K524. Carbamylation of RSA had no effect on glomerular filtration or proximal tubule uptake. These data indicate urea-mediated time-dependent carbamylation of albumin lysine K524 resulted in reduced binding to CUB7,8 and FcRn that contribute to altered albumin transport, leading to increased vascular clearance and increased liver and endothelial tissue accumulation.
Collapse
MESH Headings
- Animals
- Chromatography, Liquid
- Disease Models, Animal
- Glomerular Filtration Rate
- Histocompatibility Antigens Class I/metabolism
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/physiopathology
- Liver/metabolism
- Lysine
- Male
- Microscopy, Fluorescence, Multiphoton
- Protein Binding
- Protein Carbamylation
- Rats, Inbred Strains
- Rats, Sprague-Dawley
- Receptors, Cell Surface/metabolism
- Receptors, Fc/metabolism
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/physiopathology
- Scattering, Small Angle
- Serum Albumin/metabolism
- Tandem Mass Spectrometry
- Time Factors
- X-Ray Diffraction
- Rats
Collapse
Affiliation(s)
- Shiv Pratap S Yadav
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ruben M Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jingfu Zhao
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas
| | - Yifan Huang
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas
| | - Exing Wang
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, Texas
| | - Sudhanshu Kumar
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Silvia B Campos-Bilderback
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - George Rhodes
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas
| | - Bruce A Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark C Wagner
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
14
|
Martínez-Klimova E, Aparicio-Trejo OE, Gómez-Sierra T, Jiménez-Uribe AP, Bellido B, Pedraza-Chaverri J. Mitochondrial dysfunction and endoplasmic reticulum stress in the promotion of fibrosis in obstructive nephropathy induced by unilateral ureteral obstruction. Biofactors 2020; 46:716-733. [PMID: 32905648 DOI: 10.1002/biof.1673] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/06/2020] [Indexed: 12/18/2022]
Abstract
Obstructive nephropathy favors the progression to chronic kidney disease (CKD), a severe health problem worldwide. The unilateral ureteral obstruction (UUO) model is used to study the development of fibrosis. Impairment of renal mitochondria plays a crucial role in several types of CKD and has been strongly related to fibrosis onset. Nevertheless, in the UUO model, the impairment of mitochondria, their relationship with endoplasmic reticulum (ER) stress induction and the participation of both to induce the fibrotic process remain unclear. In this review, we summarize the current information about mitochondrial bioenergetics, redox dynamics, mitochondrial mass, and biogenesis alterations, as well as the relationship of these mitochondrial alterations with ER stress and their participation in fibrotic processes in UUO models. Early after obstruction, there is metabolic reprogramming related to mitochondrial fatty acid β-oxidation impairment, triggering lipid deposition, oxidative stress, (calcium) Ca2+ dysregulation, and a reduction in mitochondrial mass and biogenesis. Mitochondria and the ER establish a pathological feedback loop that promotes the impairment of both organelles by ER stress pathways and Ca2+ levels dysregulation. Preserving mitochondrial and ER function can prevent or at least delay the fibrotic process and loss of renal function. However, deeper understanding is still necessary for future clinically-useful therapies.
Collapse
Affiliation(s)
- Elena Martínez-Klimova
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Mexico, Mexico
| | | | - Tania Gómez-Sierra
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Mexico, Mexico
| | | | - Belen Bellido
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - José Pedraza-Chaverri
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Mexico, Mexico
| |
Collapse
|
15
|
Guan T, Fang F, Su X, Lin K, Gao Q. Silencing PEG3 inhibits renal fibrosis in a rat model of diabetic nephropathy by suppressing the NF-κB pathway. Mol Cell Endocrinol 2020; 513:110823. [PMID: 32311421 DOI: 10.1016/j.mce.2020.110823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/17/2019] [Accepted: 04/10/2020] [Indexed: 12/22/2022]
Abstract
As paternally expressed gene 3 (PEG3), which can activate NF-κB pathway, plays an important role in the development of renal fibrosis in diabetic nephropathy (DN), the present study aimed to investigate the interaction of PEG3 and the NF-κB pathway on renal fibrosis in a DN model. Following the induction of the rat model of DN, a series of experiments were used to measure serum creatinine (Scr), blood urea nitrogen (BUN), urine protein for 24 h (UP24 h), proliferation of renal fibroblasts, positive expression of PEG3, Collagen I and Collagen II protein, the activity of NF-κB, collagen fiber expression and the FSP1+ cell ratio (fibroblast marker, reflecting renal fibrosis). Silencing of PEG3 or inhibition of the NF-κB pathway decreased the levels of Scr, BUN, and UP24 h, down-regulated Collagen I protein and up-regulated Collagen II protein. These treatments also down-regulated the expression of PEG3, NF-κB, Vimentin, α-SMA, FN, caspase-3 and FSP1 and the extents of IκBα, inhibitor of kappa B (IκB) kinase β (IKKβ), and NF-κB p65 phosphorylation while that of E-cadherin was up-regulated, and the ratio of FSP1+ cells was decreased. Taken together, these results showed that silencing of PEG3 inhibited the NF-κB pathway, thereby alleviating renal fibrosis in DN, thus presenting PEG3 as a potential therapeutic target in renal fibrosis in DN.
Collapse
Affiliation(s)
- Tianjun Guan
- Department of Nephrology, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361004, PR China
| | - Fan Fang
- Department of Nephrology, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361004, PR China
| | - Xiaoxuan Su
- Department of Nephrology, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361004, PR China
| | - Kaiqiang Lin
- Department of Nephrology, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361004, PR China
| | - Qing Gao
- Department of Nephrology, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, 361004, PR China.
| |
Collapse
|
16
|
Packer M. Mechanisms Leading to Differential Hypoxia-Inducible Factor Signaling in the Diabetic Kidney: Modulation by SGLT2 Inhibitors and Hypoxia Mimetics. Am J Kidney Dis 2020; 77:280-286. [PMID: 32711072 DOI: 10.1053/j.ajkd.2020.04.016] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/26/2020] [Indexed: 12/13/2022]
Abstract
Sodium/glucose cotransporter 2 (SGLT2) inhibitors exert important renoprotective effects in the diabetic kidney, which cannot be readily explained by their actions to lower blood glucose, blood pressure, or glomerular filtration pressures. Their effects to promote erythrocytosis suggest that these drugs act on hypoxia-inducible factors (HIFs; specifically, HIF-1α and HIF-2α), which may underlie their ability to reduce the progression of nephropathy. Type 2 diabetes is characterized by renal hypoxia, oxidative and endoplasmic reticulum stress, and defective nutrient deprivation signaling, which (acting in concert) are poised to cause both activation of HIF-1α and suppression of HIF-2α. This shift in the balance of HIF-1α/HIF-2α activities promotes proinflammatory and profibrotic pathways in glomerular and renal tubular cells. SGLT2 inhibitors alleviate renal hypoxia and cellular stress and enhance nutrient deprivation signaling, which collectively may explain their actions to suppress HIF-1α and activate HIF-2α and thereby augment erythropoiesis, while muting organellar dysfunction, inflammation, and fibrosis. Cobalt chloride, a drug conventionally classified as a hypoxia mimetic, has a profile of molecular and cellular actions in the kidney that is similar to those of SGLT2 inhibitors. Therefore, many renoprotective benefits of SGLT2 inhibitors may be related to their effect to promote oxygen deprivation signaling in the diabetic kidney.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX; Imperial College, London, United Kingdom.
| |
Collapse
|
17
|
Packer M. Role of Impaired Nutrient and Oxygen Deprivation Signaling and Deficient Autophagic Flux in Diabetic CKD Development: Implications for Understanding the Effects of Sodium-Glucose Cotransporter 2-Inhibitors. J Am Soc Nephrol 2020; 31:907-919. [PMID: 32276962 DOI: 10.1681/asn.2020010010] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Growing evidence indicates that oxidative and endoplasmic reticular stress, which trigger changes in ion channels and inflammatory pathways that may undermine cellular homeostasis and survival, are critical determinants of injury in the diabetic kidney. Cells are normally able to mitigate these cellular stresses by maintaining high levels of autophagy, an intracellular lysosome-dependent degradative pathway that clears the cytoplasm of dysfunctional organelles. However, the capacity for autophagy in both podocytes and renal tubular cells is markedly impaired in type 2 diabetes, and this deficiency contributes importantly to the intensity of renal injury. The primary drivers of autophagy in states of nutrient and oxygen deprivation-sirtuin-1 (SIRT1), AMP-activated protein kinase (AMPK), and hypoxia-inducible factors (HIF-1α and HIF-2α)-can exert renoprotective effects by promoting autophagic flux and by exerting direct effects on sodium transport and inflammasome activation. Type 2 diabetes is characterized by marked suppression of SIRT1 and AMPK, leading to a diminution in autophagic flux in glomerular podocytes and renal tubules and markedly increasing their susceptibility to renal injury. Importantly, because insulin acts to depress autophagic flux, these derangements in nutrient deprivation signaling are not ameliorated by antihyperglycemic drugs that enhance insulin secretion or signaling. Metformin is an established AMPK agonist that can promote autophagy, but its effects on the course of CKD have been demonstrated only in the experimental setting. In contrast, the effects of sodium-glucose cotransporter-2 (SGLT2) inhibitors may be related primarily to enhanced SIRT1 and HIF-2α signaling; this can explain the effects of SGLT2 inhibitors to promote ketonemia and erythrocytosis and potentially underlies their actions to increase autophagy and mute inflammation in the diabetic kidney. These distinctions may contribute importantly to the consistent benefit of SGLT2 inhibitors to slow the deterioration in glomerular function and reduce the risk of ESKD in large-scale randomized clinical trials of patients with type 2 diabetes.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas .,Imperial College, London, United Kingdom
| |
Collapse
|
18
|
Maekawa H, Inagi R. Pathophysiological Role of Organelle Stress/Crosstalk in AKI-to-CKD Transition. Semin Nephrol 2019; 39:581-588. [DOI: 10.1016/j.semnephrol.2019.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|