1
|
Zhang P, Zhang W, Han Y, Yang T, Zhong J, Yun H, Fang L. Investigation of the connection between triglyceride-glucose (TyG) index and the risk of acute kidney injury in septic patients - a retrospective analysis utilizing the MIMIC-IV database. Ren Fail 2025; 47:2449199. [PMID: 39763061 PMCID: PMC11721622 DOI: 10.1080/0886022x.2024.2449199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025] Open
Abstract
The TyG index serves as a valuable tool for evaluating insulin resistance. An elevated TyG has shown a strong association with the occurrence of acute kidney injury (AKI). Nevertheless, existing literature does not address the relationship between the TyG index and acute kidney injury in patients with sepsis. Sepsis patients were identified from the MIMIC-IV database and categorized into four groups according to quadrilles of their TyG index values. The primary outcome of this study was the incidence of AKI. The relationship between the TyG index and the risk of AKI in septic patients was evaluated using Cox proportional hazards and restricted cubic spline models. Subgroup analyses were conducted to investigate the prognostic value of the TyG index in different subgroups. A total of 2,616 patients with sepsis (57% of whom were male) were included in this study. The incidence of AKI was found to be 78%. Cox proportional hazards analysis revealed a significant correlation between the TyG index and the occurrence of AKI in septic patients. Furthermore, a restricted cubic spline model revealed an approximately linear relationship between a higher TyG index and an elevated risk of AKI in septic patients. The trend of the hazard ratio (HR) remained consistent across various subgroups. These findings emphasize the reliability of the TyG index as an independent predictor for the occurrence of AKI and unfavorable renal outcomes in sepsis patients. Nevertheless, establishing a causal relationship between the two requires demonstration through larger prospective studies.
Collapse
Affiliation(s)
- Pirun Zhang
- The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Wenli Zhang
- Qingdao Mental Health Center, Qingdao, Shandong Province, China
| | - Yan Han
- The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Tong Yang
- The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jiayi Zhong
- The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Han Yun
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
- Chao En-xiang Famous Chinese Medicine Expert Inheritance Studio, Guangzhou, Guangdong Province, China
| | - Lai Fang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
- Chao En-xiang Famous Chinese Medicine Expert Inheritance Studio, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Li C, Liu X, Lv X, Zhou W, Fan G, Zhu F. The application of the triglyceride-glucose-body mass index (TyG-BMI) in predicting acute kidney injury in diabetic patients following coronary artery bypass grafting surgery. J Cardiothorac Surg 2025; 20:221. [PMID: 40281638 PMCID: PMC12023640 DOI: 10.1186/s13019-025-03455-1] [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: 07/24/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND The triglyceride-glucose-body mass index (TyG-BMI), a marker for insulin resistance, is recognized for its predictive role in cardiovascular and metabolic diseases, including kidney disease. we explored the TyG-BMI index's association with postoperative kidney injury in coronary artery bypass grafting (CABG) patients, who are at an elevated risk for such complications, underscoring its potential as a predictor for acute kidney injury (AKI). METHODS This single-center, retrospective study included 126 patients. Patients were divided into AKI and non-AKI groups postoperatively according to the KDIGO classification criteria. Univariate logistic regression was used to screen for variables with significant differences (P < 0.01), and multiple multivariate regression models were constructed to analyze independent risk factors in the multivariate regression model and to analyze the value of TyG-BMI in predicting AKI in diabetic patients after CABG. RESULTS Compared to the non-AKI group, the AKI group had statistically significant differences in preoperative fasting triglycerides, preoperative fasting glucose, preoperative and postoperative creatinine levels, ICU stay duration, and TyG-BMI levels (P < 0.05). Based on the results of univariate regression analysis, a multivariate logistic regression model A was constructed using all significant variables, and a multivariate logistic regression model 2 was constructed using significant variables other than TyG-BMI. ROC analysis showed that model 2 had better predictive performance than model 1 (AUC = 0.836 vs. 0.766). A positive correlation was observed between TyG-BMI and AKI occurrence (Spearman's correlation coefficient: R = 0.33, P = 0.00019). CONCLUSION Elevated TyG-BMI levels are closely associated with AKI in diabetic patients after CABG. TyG-BMI has potentially predictive value for AKI in diabetic patients after CABG and may play a crucial role in risk stratification in clinical practice.
Collapse
Affiliation(s)
- Chen Li
- Department of Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xiaobin Liu
- Department of Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xingping Lv
- Department of Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wei Zhou
- Department of Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Guoliang Fan
- Department of Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Feng Zhu
- Department of Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
3
|
Qin W, Nie P, Hui X, Chen F, Hu X, Shi W, Luo M, Li B. Research progress of hypoxia-inducible factor-1α and zinc in the mechanism of diabetic kidney disease. Front Pharmacol 2025; 16:1537749. [PMID: 39995420 PMCID: PMC11847805 DOI: 10.3389/fphar.2025.1537749] [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: 12/01/2024] [Accepted: 01/16/2025] [Indexed: 02/26/2025] Open
Abstract
Diabetic kidney disease is one of the common complications in diabetic patients and has gradually become an important pathogenic factor in chronic kidney disease. Therefore, studying the mechanisms of its occurrence and development is of great significance for the prevention and treatment of diabetic kidney disease. Some researchers have pointed out that there is a phenomenon of hypoxia in diabetic kidney tissue and believe that hypoxia-inducible factor-1α is closely related to the occurrence and progression of diabetic kidney disease. Additionally, the homeostasis of zinc plays a key role in the body's adaptation to hypoxic environments. However, the specific relationship among these three factors remains unclear. This article provides a detailed review of the multiple roles of hypoxia-inducible factor-1α in the pathogenesis of diabetic kidney disease, including: regulating angiogenesis, increasing the expression of erythropoietin, modulating oxidative stress through the PI3K/AKT and HIF-1α/HO-1 pathways, promoting inflammatory cell infiltration and the release of inflammatory factors to induce inflammatory responses, facilitating epithelial-mesenchymal transition, pathological angiogenesis, and promoting the release of fibrotic factors, ultimately leading to renal fibrosis. Furthermore, HIF-1α also participates in the occurrence and development of diabetic kidney disease through mechanisms such as regulating apoptosis, inducing mitochondrial autophagy, and vascular calcification. At the same time, this article clarifies the regulatory role of the trace element zinc on hypoxia-inducible factor-1α in diabetic kidney disease. This article provides references and insights for further research on the pathogenesis and progression of diabetic kidney disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Manyu Luo
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Bing Li
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
4
|
Sánchez-Cazorla E, Carrera N, García-González MÁ. HNF1B Transcription Factor: Key Regulator in Renal Physiology and Pathogenesis. Int J Mol Sci 2024; 25:10609. [PMID: 39408938 PMCID: PMC11476927 DOI: 10.3390/ijms251910609] [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: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
The HNF1B gene, located on chromosome 17q12, encodes a transcription factor essential for the development of several organs. It regulates the expression of multiple genes in renal, pancreatic, hepatic, neurological, and genitourinary tissues during prenatal and postnatal development, influencing processes such as nephrogenesis, cellular polarity, tight junction formation, cilia development, ion transport in the renal tubule, and renal metabolism. Mutations that alter the function of Hnf1b deregulate those processes, leading to various pathologies characterized by both renal and extrarenal manifestations. The main renal diseases that develop are polycystic kidney disease, hypoplastic or dysplastic kidneys, structural abnormalities, Congenital Anomalies of the Kidney and Urinary Tract (CAKUT), and electrolyte imbalances such as hyperuricemia and hypomagnesemia. Extrarenal manifestations include Maturity-Onset Diabetes of the Young (MODY), hypertransaminasemia, genital and urinary tract malformations, Autism Spectrum Disorder (ASD), and other neurodevelopmental disorders. Patients with HNF1B alterations typically carry either punctual mutations or a monoallelic microdeletion in the 17q12 region. Future research on the molecular mechanisms and genotype-phenotype correlations in HNF1B-related conditions will enhance our understanding, leading to improved clinical management, genetic counseling, monitoring, and patient care.
Collapse
Affiliation(s)
- Eloísa Sánchez-Cazorla
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
| | - Noa Carrera
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
- RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| | - Miguel Ángel García-González
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
- RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| |
Collapse
|
5
|
Lumpuy-Castillo J, Amador-Martínez I, Díaz-Rojas M, Lorenzo O, Pedraza-Chaverri J, Sánchez-Lozada LG, Aparicio-Trejo OE. Role of mitochondria in reno-cardiac diseases: A study of bioenergetics, biogenesis, and GSH signaling in disease transition. Redox Biol 2024; 76:103340. [PMID: 39250857 PMCID: PMC11407069 DOI: 10.1016/j.redox.2024.103340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are global health burdens with rising prevalence. Their bidirectional relationship with cardiovascular dysfunction, manifesting as cardio-renal syndromes (CRS) types 3 and 4, underscores the interconnectedness and interdependence of these vital organ systems. Both the kidney and the heart are critically reliant on mitochondrial function. This organelle is currently recognized as a hub in signaling pathways, with emphasis on the redox regulation mediated by glutathione (GSH). Mitochondrial dysfunction, including impaired bioenergetics, redox, and biogenesis pathways, are central to the progression of AKI to CKD and the development of CRS type 3 and 4. This review delves into the metabolic reprogramming and mitochondrial redox signaling and biogenesis alterations in AKI, CKD, and CRS. We examine the pathophysiological mechanisms involving GSH redox signaling and the AMP-activated protein kinase (AMPK)-sirtuin (SIRT)1/3-peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α) axis in these conditions. Additionally, we explore the therapeutic potential of GSH synthesis inducers in mitigating these mitochondrial dysfunctions, as well as their effects on inflammation and the progression of CKD and CRS types 3 and 4.
Collapse
Affiliation(s)
- Jairo Lumpuy-Castillo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Ciberdem, Medicine Department, Autonomous University, 28040, Madrid, Spain.
| | - Isabel Amador-Martínez
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico; Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510, Mexico City, Mexico.
| | - Miriam Díaz-Rojas
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 43210, Columbus, Ohio, USA.
| | - Oscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Ciberdem, Medicine Department, Autonomous University, 28040, Madrid, Spain.
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510, Mexico City, Mexico.
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico.
| | - Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico.
| |
Collapse
|
6
|
Qiyan Zheng, Zhang X, Guo J, Wang Y, Jiang Y, Li S, Liu YN, Liu WJ. JinChan YiShen TongLuo Formula ameliorate mitochondrial dysfunction and apoptosis in diabetic nephropathy through the HIF-1α-PINK1-Parkin pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117863. [PMID: 38325670 DOI: 10.1016/j.jep.2024.117863] [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: 11/16/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The JinChan YiShen TongLuo (JCYSTL) formula, a traditional Chinese medicine (TCM), has been used clinically for decades to treat diabetic nephropathy (DN). TCM believes that the core pathogenesis of DN is "kidney deficiency and collateral obstruction," and JCYSTL has the effect of "tonifying kidney and clearing collateral," thus alleviating the damage to kidney structure and function caused by diabetes. From the perspective of modern medicine, mitochondrial damage is an important factor in DN pathogenesis. Our study suggests that the regulation of mitophagy and mitochondrial function by JCYSTL may be one of the internal mechanisms underlying its good clinical efficacy. AIM OF THE STUDY This study aimed to investigate the mechanisms underlying the renoprotective effects of JCYSTL. MATERIALS AND METHODS Unilateral nephrectomy combined with low-dose streptozotocin intraperitoneally injected in a DN rat model and high glucose (HG) plus hypoxia-induced HK-2 cells were used to explore the effects of JCYSTL on the HIF-1α/mitophagy pathway, mitochondrial function and apoptosis. RESULTS JCYSTL treatment significantly decreased albuminuria, serum creatinine, blood urea nitrogen, and uric acid levels and increased creatinine clearance levels in DN rats. In vitro, medicated serum containing JCYSTL formula increased mitochondrial membrane potential (MMP); improved activities of mitochondrial respiratory chain complexes I, III, and IV; decreased the apoptotic cell percentage and apoptotic protein Bax expression; and increased anti-apoptotic protein Bcl-2 expression in HG/hypoxia-induced HK-2 cells. The treatment group exhibited increased accumulation of PINK1, Parkin, and LC3-II and reduced P62 levels in HG/hypoxia-induced HK-2 cells, whereas in PINK1 knockdown HK-2 cells, JCYSTL did not improve the HG/hypoxia-induced changes in Parkin, LC3-II, and P62. When mitophagy was impaired by PINK1 knockdown, the inhibitory effect of JCYSTL on Bax and its promoting effect on MMP and Bcl-2 disappeared. The JCYSTL-treated group displayed significantly higher HIF-1α expression than the model group in vivo, which was comparable to the effects of FG-4592 in DN rats. PINK1 knockdown did not affect HIF-1α accumulation in JCYSTL-treated HK-2 cells exposed to HG/hypoxia. Both JCYSTL and FG-4592 ameliorated mitochondrial morphological abnormalities and reduced the mitochondrial respiratory chain complex activity in the renal tubules of DN rats. Mitochondrial apoptosis signals in DN rats, such as increased Bax and Caspase-3 expression and apoptosis ratio, were weakened by JCYSTL or FG-4592 administration. CONCLUSION This study demonstrates that the JCYSTL formula activates PINK1/Parkin-mediated mitophagy by stabilizing HIF-1α to protect renal tubules from mitochondrial dysfunction and apoptosis in diabetic conditions, presenting a promising therapy for the treatment of DN.
Collapse
Affiliation(s)
- Qiyan Zheng
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518000, China; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China; Renal Research Institution of Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Xueqin Zhang
- Hebei University of Chinese Medicine, Hebei, 050020, China
| | - Jing Guo
- China Academy of Chinese Medicine Science, Beijing, 100700, China
| | - Yahui Wang
- Fangshan Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Yuhua Jiang
- China Academy of Chinese Medicine Science, Beijing, 100700, China
| | - Shunmin Li
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518000, China.
| | - Yu Ning Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China; Renal Research Institution of Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Wei Jing Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China; Renal Research Institution of Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
7
|
Jin Z, Zhang K. Association between triglyceride-glucose index and AKI in ICU patients based on MIMICIV database: a cross-sectional study. Ren Fail 2023; 45:2238830. [PMID: 37563796 PMCID: PMC10424620 DOI: 10.1080/0886022x.2023.2238830] [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: 04/11/2023] [Revised: 06/30/2023] [Accepted: 07/15/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Methods for early prediction of the occurrence of acute kidney injury (AKI) were limited. The relationship between triglyceride glucose index (TyG) and the incidence of acute kidney injury in ICU patients is unclear. This study aims to explore the relationship between the two. METHODS Based on their TyG index, participants from the Intensive Care Medical Information Market IV (MIMIC-IV) were divided into quartiles. A logistic regression model was constructed based on the risk of acute kidney injury as the main outcome, in order to detect a potential relationship that may exist between the TyG index and acute kidney injury in ICU patients. Finally, in order to confirm the relationship existing between the TyG index and the results, a restricted cubic spline model was used. RESULTS In total, 54,263 patients were involved in our present study, of whom 48.2% were male. The occurrence of acute kidney injury was 25.1%. An independent relationship was observed between the TyG index and an increased risk of acute kidney injury through multivariate logistic regression analysis (OR, 1.28 [95% CI 1.22-1.35] p < 0.001). Q4 (5.344-9.911) of the TyG index quartiles was independently associated with an increase in the risk of acute kidney injury (OR, 1.43 [95% CI (1.32-1.54)] p < 0.001). Through the restricted cubic spline regression model, the risk of acute kidney injury was also demonstrated to increase linearly with an increase in the TyG index. CONCLUSION The triglyceride glucose index is related to the risk of acute kidney injury in ICU patients. In the future, in order to further validate this finding, larger prospective studies are needed.
Collapse
Affiliation(s)
- Zihan Jin
- Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Kai Zhang
- The Second Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
8
|
Peng K, Xie W, Wang T, Li Y, de Dieu Habimana J, Amissah OB, Huang J, Chen Y, Ni B, Li Z. HIF-1α promotes kidney organoid vascularization and applications in disease modeling. Stem Cell Res Ther 2023; 14:336. [PMID: 37981699 PMCID: PMC10659095 DOI: 10.1186/s13287-023-03528-9] [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: 07/18/2023] [Accepted: 10/09/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Kidney organoids derived from human pluripotent stem cells (HiPSCs) hold huge applications for drug screening, disease modeling, and cell transplanting therapy. However, these applications are limited since kidney organoid cannot maintain complete morphology and function like human kidney. Kidney organoids are not well differentiated since the core of the organoid lacked oxygen, nutrition, and vasculature, which creates essential niches. Hypoxia-inducible factor-1 α (HIF-1α) serves as a critical regulator in vascularization and cell survival under hypoxia environment. Less is known about the role of HIF-1α in kidney organoids in this regard. This study tried to investigate the effect of HIF-1α in kidney organoid vascularization and related disease modeling. METHODS For the vascularization study, kidney organoids were generated from human induced pluripotent stem cells. We overexpressed HIF-1α via plasmid transfection or treated DMOG (Dimethyloxallyl Glycine, an agent for HIF-1α stabilization and accumulation) in kidney progenitor cells to detect the endothelium. For the disease modeling study, we treated kidney organoid with cisplatin under hypoxia environment, with additional HIF-1α transfection. RESULT HIF-1α overexpression elicited kidney organoid vascularization. The endothelial cells and angiotool analysis parameters were increased in HIF-1α plasmid-transfected and DMOG-treated organoids. These angiogenesis processes were partially blocked by VEGFR inhibitors, semaxanib or axitinib. Cisplatin-induced kidney injury (Cleaved caspase 3) was protected by HIF-1α through the upregulation of CD31 and SOD2. CONCLUSION We demonstrated that HIF-1α elicited the process of kidney organoid vascularization and protected against cisplatin-induced kidney organoid injury in hypoxia environment.
Collapse
Affiliation(s)
- Kexin Peng
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Wanqin Xie
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Tingting Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Yamei Li
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Jean de Dieu Habimana
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Obed Boadi Amissah
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Jufang Huang
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yong Chen
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Bin Ni
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.
| | - Zhiyuan Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, China.
- GZMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China.
- GIBH-CUHK Joint Research Laboratory On Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou, China.
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China.
| |
Collapse
|
9
|
Louis F, Rioult D, Rocher B, Gaillet V, Delahaut L, Paris-Palacios S, David E. Dreissena polymorpha responses under thermal and hypoxic stress: New insights in the tolerance of this freshwater sentinel species. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 260:106586. [PMID: 37247577 DOI: 10.1016/j.aquatox.2023.106586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 05/31/2023]
Abstract
Dreissena polymorpha is a sentinel freshwater mussel providing key functional ecosystemic services like nutrient recycling and suspended matter filtration. Global warming and especially extreme events imply rapid fluctuations of environmental parameters that sessile organisms could not escape. The increase occurrence of heat waves and the subsequent expansion of hypoxic areas could challenge the survival of mussels. This study provided a deeper knowledge of energy management and cellular function during thermal (+15 °C) or hypoxic (30% of dissolved oxygen saturation) stress for 7 days. A potential metabolic rate depression was highlighted in D. polymorpha under hypoxia through a decline in the mitochondrial activity and a constant AMP content over time. A contrasted pattern of response was observed in thermal-stressed mussels between 24 h and 7 days of exposure. A global increase of metabolic activity was noticed in mussels after 24 h while a return to control level was noticed at the end of the experiment. Although D. polymorpha is considered as a temperature tolerant species, a significant increase of ADP:ATP ratio, related to a decrease of mitochondrial activity and density, suggested an overwhelming of organisms. This study pointed to the importance of considering time of exposure to natural factor variations in tolerance window of organisms in a long-term changing environment. The apparent short-term tolerance of D. polymorpha could hide much more deleterious consequences, i.e. mortality, if abiotic stresses persist, as suggested by climate change models.
Collapse
Affiliation(s)
- Fanny Louis
- Université de Reims Champagne-Ardenne (URCA), UMR-I 02 SEBIO, UFR Sciences Exactes et Naturelles, Campus Moulin de la Housse, BP 1039, 51687 Reims Cedex 02, France
| | - Damien Rioult
- Université de Reims Champagne-Ardenne (URCA), UMR-I 02 SEBIO, UFR Sciences Exactes et Naturelles, Campus Moulin de la Housse, BP 1039, 51687 Reims Cedex 02, France; Plateau Technique Mobile de Cytométrie Environnementale MOBICYTE, Université de Reims Champagne-Ardenne/INERIS, 51687 Reims Cedex 02, France
| | - Béatrice Rocher
- Université Le Havre Normandie (ULHN), UMR-I 02 SEBIO, FR CNRS 3730 SCALE, 25 rue Philippe Le Bon, Le Havre, 76600, France
| | - Véronique Gaillet
- Université de Reims Champagne-Ardenne (URCA), UMR-I 02 SEBIO, UFR Sciences Exactes et Naturelles, Campus Moulin de la Housse, BP 1039, 51687 Reims Cedex 02, France
| | - Laurence Delahaut
- Université de Reims Champagne-Ardenne (URCA), UMR-I 02 SEBIO, UFR Sciences Exactes et Naturelles, Campus Moulin de la Housse, BP 1039, 51687 Reims Cedex 02, France
| | - Séverine Paris-Palacios
- Université de Reims Champagne-Ardenne (URCA), UMR-I 02 SEBIO, UFR Sciences Exactes et Naturelles, Campus Moulin de la Housse, BP 1039, 51687 Reims Cedex 02, France
| | - Elise David
- Université de Reims Champagne-Ardenne (URCA), UMR-I 02 SEBIO, UFR Sciences Exactes et Naturelles, Campus Moulin de la Housse, BP 1039, 51687 Reims Cedex 02, France.
| |
Collapse
|
10
|
Łagosz P, Biegus J, Urban S, Zymliński R. Renal Assessment in Acute Cardiorenal Syndrome. Biomolecules 2023; 13:biom13020239. [PMID: 36830608 PMCID: PMC9953721 DOI: 10.3390/biom13020239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 01/28/2023] Open
Abstract
Cardiorenal syndrome (CRS) is a complex, heterogeneous spectrum of symptoms that has kept cardiologists awake for decades. The heart failure (HF) population being burdened with multimorbidity poses diagnostic and therapeutic challenges even for experienced clinicians. Adding deteriorated renal function to the equation, which is one of the strongest predictors of adverse outcome, we measure ourselves against possibly the biggest problem in modern cardiology. With the rapid development of new renal assessment methods, we can treat CRS more effectively than ever. The presented review focuses on explaining the pathophysiology, recent advances and current practices of monitoring renal function in patients with acute CRS. Understanding the dynamic interaction between the heart and the kidney may improve patient care and support the selection of an effective and nephroprotective treatment strategy.
Collapse
Affiliation(s)
- Piotr Łagosz
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Institute of Heart Diseases, University Clinical Hospital, 50-556 Wroclaw, Poland
- Correspondence:
| | - Jan Biegus
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Institute of Heart Diseases, University Clinical Hospital, 50-556 Wroclaw, Poland
| | - Szymon Urban
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Robert Zymliński
- Institute of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Institute of Heart Diseases, University Clinical Hospital, 50-556 Wroclaw, Poland
| |
Collapse
|
11
|
Li ZL, Wang B, Wen Y, Wu QL, Lv LL, Liu BC. Disturbance of Hypoxia Response and Its Implications in Kidney Diseases. Antioxid Redox Signal 2022; 37:936-955. [PMID: 35044244 DOI: 10.1089/ars.2021.0271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: The disturbance of the hypoxia response system is closely related to human diseases, because it is essential for the maintenance of homeostasis. Given the significant role of the hypoxia response system in human health, therapeutic applications targeting prolyl hydroxylase-hypoxia-inducible factor (HIF) signaling have been attempted. Thus, systemically reviewing the hypoxia response-based therapeutic strategies is of great significance. Recent Advances: Disturbance of the hypoxia response is a characteristic feature of various diseases. Targeting the hypoxia response system is, thus, a promising therapeutic strategy. Interestingly, several compounds and drugs are currently under clinical trials, and some have already been approved for use in the treatment of certain human diseases. Critical Issues: We summarize the molecular mechanisms of the hypoxia response system and address the potential therapeutic implications in kidney diseases. Given that the effects of hypoxia response in kidney diseases are likely to depend on the pathological context, specific cell types, and the differences in the activation pattern of HIF isoforms, the precise application is critical for the treatment of kidney diseases. Although HIF-PHIs (HIF-PHD inhibitors) have been proven to be effective and well tolerated in chronic kidney disease patients with anemia, the potential on-target consequence of HIF activation and some outstanding questions warrant further consideration. Future Direction: The mechanism of the hypoxia response system disturbance remains unclear. Elucidation of the molecular mechanism of hypoxia response and its precise effects on kidney diseases warrants clarification. Considering the complexity of the hypoxia response system and multiple biological processes controlled by HIF signaling, the development of more specific inhibitors is highly warranted. Antioxid. Redox Signal. 37, 936-955.
Collapse
Affiliation(s)
- Zuo-Lin Li
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - Bin Wang
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yi Wen
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - Qiu-Li Wu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
12
|
Locatelli F, Minutolo R, De Nicola L, Del Vecchio L. Evolving Strategies in the Treatment of Anaemia in Chronic Kidney Disease: The HIF-Prolyl Hydroxylase Inhibitors. Drugs 2022; 82:1565-1589. [PMID: 36350500 PMCID: PMC9645314 DOI: 10.1007/s40265-022-01783-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2022] [Indexed: 11/11/2022]
Abstract
Chronic kidney disease (CKD) affects approximately 10% of the worldwide population; anaemia is a frequent complication. Inadequate erythropoietin production and absolute or functional iron deficiency are the major causes. Accordingly, the current treatment is based on iron and erythropoiesis stimulating agents (ESAs). Available therapy has dramatically improved the management of anaemia and the quality of life. However, safety concerns were raised over ESA use, especially when aiming to reach near-to-normal haemoglobin levels with high doses. Moreover, many patients show hypo-responsiveness to ESA. Hypoxia-inducible factor (HIF) prolyl hydroxylase domain (PHD) inhibitors (HIF-PHIs) were developed for the oral treatment of anaemia in CKD to overcome these concerns. They simulate the body's exposure to moderate hypoxia, stimulating the production of endogenous erythropoietin. Some molecules are already approved for clinical use in some countries. Data from clinical trials showed non-inferiority in anaemia correction compared to ESA or superiority for placebo. Hypoxia-inducible factor-prolyl hydroxylase domain inhibitors may also have additional advantages in inflamed patients, improving iron utilisation and mobilisation and decreasing LDL-cholesterol. Overall, non-inferiority was also shown in major cardiovascular events, except for one molecule in the non-dialysis population. This was an unexpected finding, considering the lower erythropoietin levels reached using these drugs due to their peculiar mechanism of action. More data and longer follow-ups are necessary to better clarifying safety issues and further investigate the variety of pathways activated by HIF, which could have either positive or negative effects and could differentiate HIF-PHIs from ESAs.
Collapse
Affiliation(s)
- Francesco Locatelli
- Past Director of the Department of Nephrology and Dialysis, Alessandro Manzoni Hospital, via Fratelli Cairoli 60, 23900, Lecco, Italy.
| | - Roberto Minutolo
- Nephrology and Dialysis Unit, Department of Advanced Medical and Surgical Sciences, University L. Vanvitelli, Naples, Italy
| | - Luca De Nicola
- Nephrology and Dialysis Unit, Department of Advanced Medical and Surgical Sciences, University L. Vanvitelli, Naples, Italy
| | - Lucia Del Vecchio
- Department of Nephrology and Dialysis, Sant' Anna Hospital, ASST Lariana, Como, Italy
| |
Collapse
|
13
|
Hotait ZS, Lo Cascio JN, Choos END, Shepard BD. The sugar daddy: the role of the renal proximal tubule in glucose homeostasis. Am J Physiol Cell Physiol 2022; 323:C791-C803. [PMID: 35912988 PMCID: PMC9448277 DOI: 10.1152/ajpcell.00225.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 11/22/2022]
Abstract
Renal blood flow represents >20% of total cardiac output and with this comes the great responsibility of maintaining homeostasis through the intricate regulation of solute handling. Through the processes of filtration, reabsorption, and secretion, the kidneys ensure that solutes and other small molecules are either returned to circulation, catabolized within renal epithelial cells, or excreted through the process of urination. Although this occurs throughout the renal nephron, one segment is tasked with the bulk of solute reabsorption-the proximal tubule. Among others, the renal proximal tubule is entirely responsible for the reabsorption of glucose, a critical source of energy that fuels the body. In addition, it is the only other site of gluconeogenesis outside of the liver. When these processes go awry, pathophysiological conditions such as diabetes and acidosis result. In this review, we highlight the recent advances made in understanding these processes that occur within the renal proximal tubule. We focus on the physiological mechanisms at play regarding glucose reabsorption and glucose metabolism, emphasize the conditions that occur under diseased states, and explore the emerging class of therapeutics that are responsible for restoring homeostasis.
Collapse
Affiliation(s)
- Zahraa S Hotait
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Julia N Lo Cascio
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Elijah N D Choos
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Blythe D Shepard
- Department of Human Science, Georgetown University, Washington, District of Columbia
| |
Collapse
|
14
|
A bioinspired carbon monoxide delivery system prevents acute kidney injury and the progression to chronic kidney disease. Redox Biol 2022; 54:102371. [PMID: 35763935 PMCID: PMC9241064 DOI: 10.1016/j.redox.2022.102371] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 11/20/2022] Open
Abstract
Renal ischemia-reperfusion (IR)-induced tissue hypoxia causes impaired energy metabolism and oxidative stress. These conditions lead to tubular cell damage, which is a cause of acute kidney injury (AKI) and AKI to chronic kidney disease (CKD). Three key molecules, i.e., hypoxia-inducible factor-1α (HIF-1α), AMP-activated protein kinase (AMPK), and nuclear factor E2-related factor 2 (Nrf2), have the potential to protect tubular cells from these disorders. Although carbon monoxide (CO) can comprehensively induce these three molecules via the action of mitochondrial reactive oxygen species (mtROS), the issue of whether CO induces these molecules in tubular cells remains unclear. Herein, we report that CO-enriched red blood cells (CO-RBC) cell therapy, the inspiration for which is the in vivo CO delivery system, exerts a renoprotective effect on hypoxia-induced tubular cell damage via the upregulation of the above molecules. Experiments using a mitochondria-specific antioxidant provide evidence to show that CO-driven mtROS partially contributes to the upregulation of the aforementioned molecules in tubular cells. CO-RBC ameliorates the pathological conditions of IR-induced AKI model mice via activation of these molecules. CO-RBC also prevents renal fibrosis via the suppression of epithelial mesenchymal transition and transforming growth factor-β1 secretion in an IR-induced AKI to CKD model mice. In conclusion, our results confirm that the bioinspired CO delivery system prevents the pathological conditions of both AKI and AKI to CKD via the amelioration of hypoxia inducible tubular cell damage, thereby making it an effective cell therapy for treating the progression to CKD.
Collapse
|
15
|
Li QY, Liu F, Tang X, Fu H, Mao J. Renoprotective Role of Hypoxia-Inducible Factors and the Mechanism. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:44-56. [PMID: 35224006 PMCID: PMC8820168 DOI: 10.1159/000520141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/09/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND The kidney requires abundant blood supply, and oxygen is transmitted by diffusion through blood vessels. Most physiological metabolism of the kidney depends on oxygen, so it is very sensitive to oxygen. An increasing pool of evidence suggests that hypoxia is involved in almost all acute and chronic kidney diseases (CKDs). Vascular damage, tubular injury, and fibrosis are the main pathologies associated during hypoxia. Hypoxia-inducible factors (HIFs) are the main mediators during hypoxia, but their functions remain controversial. This article reviewed recent studies and described its mechanisms on renoprotection. SUMMARY HIF is degraded rapidly during under normal oxygen. But under hypoxia, HIFs accumulate and many target genes are regulated by HIFs. Homeostasis during injury is maintained through these genes. Pretreatment of HIF can protect the kidney from acute hypoxia and can improve repair, but HIF's role in CKD and in renal tumor is still controversial. Due to its mechanism in kidney disease, many drugs toward HIFs are widely researched, even some of which have been used in clinical or in clinical research. KEY MESSAGES In this review, we described the known physiological mechanisms, target genes, and renal protective roles of HIFs, and we discussed several drugs that are researched due to such renal protective roles.
Collapse
|
16
|
Mogi M, Higashi Y, Bokuda K, Ichihara A, Nagata D, Tanaka A, Node K, Nozato Y, Yamamoto K, Sugimoto K, Shibata H, Hoshide S, Nishizawa H, Kario K. Annual reports on hypertension research 2020. Hypertens Res 2022; 45:15-31. [PMID: 34650193 DOI: 10.1038/s41440-021-00766-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
In 2020, 199 papers were published in Hypertension Research. Many excellent papers have contributed to progress in research on hypertension. Here, our editorial members have summarized eleven topics from published work and discussed current topics in depth. We hope you enjoy our special feature, Annual Reports on Hypertension Research.
Collapse
Affiliation(s)
- Masaki Mogi
- Deparment of Pharmacology, Ehime University Graduate School of Medicine, Tohon, Ehime, Japan.
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Hiroshima, Japan.,Divivsion of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Hiroshima, Japan
| | - Kanako Bokuda
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Atsuhiro Ichihara
- Department of Endocrinology and Hypertension, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Daisuke Nagata
- Division of Nephrology, Department of Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Atsushi Tanaka
- Department of Cardiovascular Medicine, Saga University, Saga, Saga, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Saga, Japan
| | - Yoichi Nozato
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ken Sugimoto
- General and Geriatric Medicine, Kawasaki Medical University, Okayama, Okayama, Japan
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Satoshi Hoshide
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| |
Collapse
|
17
|
Dizin E, Olivier V, Roth I, Sassi A, Arnoux G, Ramakrishnan S, Morel S, Kwak BR, Loffing J, Hummler E, Wenger RH, Frew IJ, Feraille E. Activation of the Hypoxia-Inducible Factor Pathway Inhibits Epithelial Sodium Channel-Mediated Sodium Transport in Collecting Duct Principal Cells. J Am Soc Nephrol 2021; 32:3130-3145. [PMID: 34615708 PMCID: PMC8638392 DOI: 10.1681/asn.2021010046] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 08/18/2021] [Accepted: 09/05/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Active sodium reabsorption is the major factor influencing renal oxygen consumption and production of reactive oxygen species (ROS). Increased sodium reabsorption uses more oxygen, which may worsen medullary hypoxia and produce more ROS via enhanced mitochondrial ATP synthesis. Both mechanisms may activate the hypoxia-inducible factor (HIF) pathway. Because the collecting duct is exposed to low oxygen pressure and variations of active sodium transport, we assessed whether the HIF pathway controls epithelial sodium channel (ENaC)-dependent sodium transport. METHODS We investigated HIF's effect on ENaC expression in mpkCCD cl4 cells (a model of collecting duct principal cells) using real-time PCR and western blot and ENaC activity by measuring amiloride-sensitive current. We also assessed the effect of hypoxia and sodium intake on abundance of kidney sodium transporters in wild-type and inducible kidney tubule-specific Hif1α knockout mice. RESULTS In cultured cells, activation of the HIF pathway by dimethyloxalylglycine or hypoxia inhibited sodium transport and decreased expression of β ENaC and γ ENaC, as well as of Na,K-ATPase. HIF1 α silencing increased β ENaC and γ ENaC expression and stimulated sodium transport. A constitutively active mutant of HIF1 α produced the opposite effect. Aldosterone and inhibition of the mitochondrial respiratory chain slowly activated the HIF pathway, suggesting that ROS may also activate HIF. Decreased γ ENaC abundance induced by hypoxia in normal mice was abolished in Hif1α knockout mice. Similarly, Hif1α knockout led to increased γ ENaC abundance under high sodium intake. CONCLUSIONS This study reveals that γ ENaC expression and activity are physiologically controlled by the HIF pathway, which may represent a negative feedback mechanism to preserve oxygenation and/or prevent excessive ROS generation under increased sodium transport.
Collapse
Affiliation(s)
- Eva Dizin
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, Geneva, Switzerland,National Centre of Competence in Research “Kidney.CH”, Switzerland
| | - Valérie Olivier
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, Geneva, Switzerland,National Centre of Competence in Research “Kidney.CH”, Switzerland
| | - Isabelle Roth
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, Geneva, Switzerland,National Centre of Competence in Research “Kidney.CH”, Switzerland
| | - Ali Sassi
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, Geneva, Switzerland,National Centre of Competence in Research “Kidney.CH”, Switzerland
| | - Grégoire Arnoux
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, Geneva, Switzerland,National Centre of Competence in Research “Kidney.CH”, Switzerland
| | - Suresh Ramakrishnan
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, Geneva, Switzerland,National Centre of Competence in Research “Kidney.CH”, Switzerland
| | - Sandrine Morel
- Department of Pathology and Immunology, University of Geneva, CMU, Geneva, Switzerland
| | - Brenda R. Kwak
- Department of Pathology and Immunology, University of Geneva, CMU, Geneva, Switzerland
| | - Johannes Loffing
- National Centre of Competence in Research “Kidney.CH”, Switzerland,Institute of Anatomy, University of Zürich, Zürich, Switzerland
| | - Edith Hummler
- National Centre of Competence in Research “Kidney.CH”, Switzerland,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Roland H. Wenger
- National Centre of Competence in Research “Kidney.CH”, Switzerland,Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Ian J. Frew
- Department of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Centre - University of Freiburg, Freiburg, Germany
| | - Eric Feraille
- Department of Cellular Physiology and Metabolism, University of Geneva, CMU, Geneva, Switzerland,National Centre of Competence in Research “Kidney.CH”, Switzerland
| |
Collapse
|
18
|
Emery JM, Ortiz RM. Mitofusin 2: A link between mitochondrial function and substrate metabolism? Mitochondrion 2021; 61:125-137. [PMID: 34536562 DOI: 10.1016/j.mito.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/22/2021] [Accepted: 09/13/2021] [Indexed: 01/18/2023]
Abstract
Mitochondria are dynamic, interactive organelles that connect cellular signaling and whole-cell homeostasis. This "mitochatting" allows the cell to receive information about the mitochondria's condition before accommodating energy demands. Mitofusin 2 (Mfn2), an outer mitochondrial membrane fusion protein specializes in mediating mitochondrial homeostasis. Early studies defined the biological significance of Mfn2, while latter studies highlighted its role in substrate metabolism. However, determining Mfn2 potential to contribute to energy homeostasis needs study. This review summarizes current literature on mitochondrial metabolic processes, dynamics, and evidence of interactions among Mfn2 and regulatory processes that may link Mfn2's role in maintaining mitochondrial function and substrate metabolism.
Collapse
Affiliation(s)
- Janna M Emery
- Department of Molecular and Cellular Biology, School of Natural Sciences, University of California, Merced, United States.
| | - Rudy M Ortiz
- Department of Molecular and Cellular Biology, School of Natural Sciences, University of California, Merced, United States
| |
Collapse
|
19
|
Zhang N, Zheng Q, Wang Y, Lin J, Wang H, Liu R, Yan M, Chen X, Yang J, Chen X. Renoprotective Effect of the Recombinant Anti-IL-6R Fusion Proteins by Inhibiting JAK2/STAT3 Signaling Pathway in Diabetic Nephropathy. Front Pharmacol 2021; 12:681424. [PMID: 34054555 PMCID: PMC8155588 DOI: 10.3389/fphar.2021.681424] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy the main reason for end stage renal disease is a common microvascular complication in patients with type 1 and type 2 diabetes. The interleukin-6 (IL-6), acting as a pleiotropic cytokine, play key roles in main autoimmune disorders. The recombinant anti-IL-6R fusion proteins (VHH-0031) constructed and obtained in our lab is a dual target-directed single domain-based fusion protein against the interleukin-6 receptor. This study aims to explore the renoprotective effect of VHH-0031 in diabetic nephropathy. VHH-0031 treatment alleviated renal inflammation, morphologic injury and renal insufficiency in both Goto-Kakizaki rats and STZ-induced Sprague Dawley rats. These renoprotective effects of VHH-0031 are associated with alleviating inflammation and suppression of the JAK2/STAT3 signaling pathway. The mesangial cells treated with VHH-0031 exhibited anti-proliferation, anti-inflammation and inactivation of JAK2/STAT3 pathway under high glucose condition. In conclusion, this study demonstrates that VHH-0031 exhibited a potent protective effect in kidney of diabetic rats and its mechanism may be concerned with the inhibition of the IL-6R/JAK2/STAT3 pathway of glomerular mesangial cells.
Collapse
Affiliation(s)
- Nanwen Zhang
- School of Pharmacy, Department of Pharmacology, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Qingmei Zheng
- School of Pharmacy, Department of Bioengineering and Biopharmaceutics, Fujian Medical University, Fuzhou, China
| | - Yaduan Wang
- School of Pharmacy, Department of Bioengineering and Biopharmaceutics, Fujian Medical University, Fuzhou, China
| | - Juan Lin
- School of Pharmacy, Department of Bioengineering and Biopharmaceutics, Fujian Medical University, Fuzhou, China
| | - He Wang
- School of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Rui Liu
- School of Pharmacy, Department of Bioengineering and Biopharmaceutics, Fujian Medical University, Fuzhou, China
| | - Mengru Yan
- School of Pharmacy, Department of Bioengineering and Biopharmaceutics, Fujian Medical University, Fuzhou, China
| | - Xiaofeng Chen
- Rehabilitation Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Juhua Yang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China.,School of Pharmacy, Department of Bioengineering and Biopharmaceutics, Fujian Medical University, Fuzhou, China
| | - Xiaole Chen
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China.,School of Pharmacy, Department of Bioengineering and Biopharmaceutics, Fujian Medical University, Fuzhou, China
| |
Collapse
|
20
|
Zou J, Yang J, Zhu X, Zhong J, Elshaer A, Matsusaka T, Pastan I, Haase VH, Yang HC, Fogo AB. Stabilization of hypoxia-inducible factor ameliorates glomerular injury sensitization after tubulointerstitial injury. Kidney Int 2020; 99:620-631. [PMID: 33137336 DOI: 10.1016/j.kint.2020.09.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022]
Abstract
Previously, we found that mild tubulointerstitial injury sensitizes glomeruli to subsequent injury. Here, we evaluated whether stabilization of hypoxia-inducible factor-α (HIF-α), a key regulator of tissue response to hypoxia, ameliorates tubulointerstitial injury and impact on subsequent glomerular injury. Nep25 mice, which express the human CD25 receptor on podocytes under control of the nephrin promotor and develop glomerulosclerosis when a specific toxin is administered were used. Tubulointerstitial injury, evident by week two, was induced by folic acid, and mice were treated with an HIF stabilizer, dimethyloxalylglycine or vehicle from week three to six. Uninephrectomy at week six assessed tubulointerstitial fibrosis. Glomerular injury was induced by podocyte toxin at week seven, and mice were sacrificed ten days later. At week six tubular injury markers normalized but with patchy collagen I and interstitial fibrosis. Pimonidazole staining, a hypoxia marker, was increased by folic acid treatment compared to vehicle while dimethyloxalylglycine stimulated HIF-2α expression and attenuated tubulointerstitial hypoxia. The hematocrit was increased by dimethyloxalylglycine along with downstream effectors of HIF. Tubular epithelial cell injury, inflammation and interstitial fibrosis were improved after dimethyloxalylglycine, with further reduced mortality, interstitial fibrosis, and glomerulosclerosis induced by specific podocyte injury. Thus, our findings indicate that hypoxia contributes to tubular injury and consequent sensitization of glomeruli to injury. Hence, restoring HIFs may blunt this adverse crosstalk of tubules to glomeruli.
Collapse
Affiliation(s)
- Jun Zou
- Division of Nephrology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jaewon Yang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Division of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Gangwon, South Korea
| | - Xiaoye Zhu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Division of Nephrology, Huashan Hospital, Wudan University, Shanghai, China
| | - Jianyong Zhong
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ahmed Elshaer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Taiji Matsusaka
- Institute of Medical Science, Tokai University, Isehara, Japan
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Volker H Haase
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Medicine and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Hai-Chun Yang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Agnes B Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
21
|
Jiang N, Zhao H, Han Y, Li L, Xiong S, Zeng L, Xiao Y, Wei L, Xiong X, Gao P, Yang M, Liu Y, Sun L. HIF-1α ameliorates tubular injury in diabetic nephropathy via HO-1-mediated control of mitochondrial dynamics. Cell Prolif 2020; 53:e12909. [PMID: 32975326 PMCID: PMC7653251 DOI: 10.1111/cpr.12909] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/19/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022] Open
Abstract
Objectives In diabetic nephropathy (DN), hypoxia‐inducible factor‐1α (HIF‐1α) activation in tubular cells plays an important protective role against kidney injury. The effects may occur via the target genes of HIF‐1α, such as haem oxygenase‐1 (HO‐1), but the exact mechanisms are incompletely understood. Materials and methods Mice with proximal tubule‐specific knockout of HIF‐1α (PT‐HIF‐1α−/− mice) were generated, and diabetes was induced in these mice by streptozotocin (STZ) injection. In addition, to mimic a hypoxic state, cobaltous chloride (CoCl2) was applied to HK‐2 cells. Results Our study first verified that conditional knockout of HIF‐1α worsened tubular injury in DN; additionally, aggravated kidney dysfunction, renal histopathological alterations, mitochondrial fragmentation, ROS accumulation and apoptosis were observed in diabetic PT‐HIF‐1α−/− mice. In vitro study showed that compared to control group, HK‐2 cells cultured under hypoxic ambiance displayed increased mitochondrial fragmentation, ROS production, mitochondrial membrane potential loss and apoptosis. These increases were reversed by overexpression of HIF‐1α or treatment with a HO‐1 agonist. Importantly, cotreatment with a HIF‐1α inhibitor and a HO‐1 agonist rescued the HK‐2 cells from the negative impacts of the HIF‐1α inhibitor. Conclusions These data revealed that HIF‐1α exerted a protective effect against tubular injury in DN, which could be mediated via modulation of mitochondrial dynamics through HO‐1 upregulation.
Collapse
Affiliation(s)
- Na Jiang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Hao Zhao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Li Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Shan Xiong
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Lingfeng Zeng
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Ying Xiao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Ling Wei
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Xiaofen Xiong
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Peng Gao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| |
Collapse
|
22
|
Hypoxia-mediated regulation of mitochondrial transcription factors in renal epithelial cells: implications for hypertensive renal physiology. Hypertens Res 2020; 44:154-167. [PMID: 32917968 DOI: 10.1038/s41440-020-00539-4] [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: 06/15/2020] [Revised: 07/19/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023]
Abstract
Kidneys have a high resting metabolic rate and low partial pressure of oxygen due to enhanced mitochondrial oxygen consumption and ATP production needed for active solute transport. Heightened mitochondrial activity leads to progressively increasing hypoxia from the renal cortex to the renal medulla. Renal hypoxia is prominent in hypertensive rats due to increased sodium reabsorption within the nephrons, which demands higher energy production by oxidative phosphorylation (OXPHOS). Consequently, spontaneously hypertensive rats (SHR) display greater oxygen deficiency (hypoxia) than normotensive Wistar Kyoto rats (WKY). Here, we sought to investigate the expression of key proteins for mitochondrial biogenesis in SHR and WKY, and study the regulation of mitochondrial transcription factors (mtTFs) under in vitro hypoxic conditions in renal epithelial cells. We report that renal expressions of hypoxia-inducible factor-1-alpha (HIF-1α), peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α), mtTFs, and OXPHOS proteins are elevated in SHR compared to WKY. In addition, our experiments in cultured kidney cells demonstrate that acute hypoxia augments the expression of these genes. Furthermore, we show that the transcripts of HIF-1α and mtTFs are positively correlated in various human tissues. We reveal, for the first time to our knowledge, that HIF-1α transactivates mtTF genes by direct interaction with their promoters in rat kidney epithelial cells (NRK-52E) under acute hypoxia. Concomitant increases in the mitochondrial DNA and RNA, and OXPHOS proteins are observed. Taken together, this study suggests that hypoxia within the renal epithelial cells may enhance mitochondrial function to meet the energy demand in proximal tubular cells during prehypertensive stages in kidneys of young SHR.
Collapse
|
23
|
Mechanism of progression of diabetic kidney disease mediated by podocyte mitochondrial injury. Mol Biol Rep 2020; 47:8023-8035. [PMID: 32918716 DOI: 10.1007/s11033-020-05749-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022]
Abstract
Diabetic kidney disease (DKD) is an important diabetic microvascular complication, which has become the main cause of end-stage renal disease (ESRD) all over the world. It is of great significance to find effective therapeutic targets and improve the prognosis of the disease. Traditionally, it is believed that the activation of the renin-angiotensin-aldosterone system (RAAS) is the main reason for the progression of DKD, but with the progress of research, it is known that the production of proteinuria in patients with DKD is also related to podocyte injury and loss. Many studies have shown that mitochondrial dysfunction in podocytes plays an important role in the occurrence and development of DKD, and oxidative stress is also the main pathway and common hub of diabetes to the occurrence and development of microvascular and macrovascular complications. Thus, the occurrence and progression of DKD is correlated with not only the activation of the RAAS, but also the damage of mitochondria, oxidative stress, and inflammatory mediators. Besides, diabetes-related metabolic disorders can also cause abnormalities in mitochondrial dynamics, autophagy and cellular signal transduction, which are intertwined in a complex way. Therefore, in this review, we mainly explore the mechanism and the latest research progress of podocyte mitochondria in DKD and summarize the main signal pathways involved in them. Thus, it provides feasible clinical application and future research suggestions for the prevention and treatment of DKD, which has important practical significance for the later treatment of patients with DKD.
Collapse
|
24
|
Aparicio-Trejo OE, Rojas-Morales P, Avila-Rojas SH, León-Contreras JC, Hernández-Pando R, Jiménez-Uribe AP, Prieto-Carrasco R, Sánchez-Lozada LG, Pedraza-Chaverri J, Tapia E. Temporal Alterations in Mitochondrial β-Oxidation and Oxidative Stress Aggravate Chronic Kidney Disease Development in 5/6 Nephrectomy Induced Renal Damage. Int J Mol Sci 2020; 21:ijms21186512. [PMID: 32899919 PMCID: PMC7555424 DOI: 10.3390/ijms21186512] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/17/2022] Open
Abstract
Five-sixths nephrectomy (5/6Nx) model is widely used for studying the mechanisms involved in chronic kidney disease (CKD) progression, a kidney pathology that has increased dramatically in recent years. Mitochondrial impairment is a key mechanism that aggravates CKD progression; however, the information on mitochondrial bioenergetics and redox alterations along a time course in a 5/6Nx model is still limited and in some cases contradictory. Therefore, we performed for the first time a time-course study of mitochondrial alterations by high-resolution respirometry in the 5/6Nx model. Our results show a decrease in mitochondrial β-oxidation at early times, as well as a permanent impairment in adenosine triphosphate (ATP) production in CI-linked respiration, a permanent oxidative state in mitochondria and decoupling of these organelles. These pathological alterations are linked to the early decrease in complex I and ATP synthase activities and to the further decrease in complex III activity. Therefore, our results may suggest that mitochondrial bioenergetics impairment is an early event in renal damage, whose persistence in time aggravates CKD development in the 5/6Nx model.
Collapse
Affiliation(s)
- Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Pathophysiology, National Institute of Cardiology “Ignacio Chávez”, Mexico City 14080, Mexico; (O.E.A.-T.); (P.R.-M.); (L.G.S.-L.)
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (S.H.A.-R.); (A.P.J.-U.); (R.P.-C.); (J.P.-C.)
| | - Pedro Rojas-Morales
- Department of Cardio-Renal Pathophysiology, National Institute of Cardiology “Ignacio Chávez”, Mexico City 14080, Mexico; (O.E.A.-T.); (P.R.-M.); (L.G.S.-L.)
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (S.H.A.-R.); (A.P.J.-U.); (R.P.-C.); (J.P.-C.)
| | - Sabino Hazael Avila-Rojas
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (S.H.A.-R.); (A.P.J.-U.); (R.P.-C.); (J.P.-C.)
| | - Juan Carlos León-Contreras
- Experimental Pathology Section, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14000, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - Rogelio Hernández-Pando
- Experimental Pathology Section, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14000, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - Alexis Paulina Jiménez-Uribe
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (S.H.A.-R.); (A.P.J.-U.); (R.P.-C.); (J.P.-C.)
| | - Rodrigo Prieto-Carrasco
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (S.H.A.-R.); (A.P.J.-U.); (R.P.-C.); (J.P.-C.)
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Pathophysiology, National Institute of Cardiology “Ignacio Chávez”, Mexico City 14080, Mexico; (O.E.A.-T.); (P.R.-M.); (L.G.S.-L.)
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico; (S.H.A.-R.); (A.P.J.-U.); (R.P.-C.); (J.P.-C.)
| | - Edilia Tapia
- Department of Cardio-Renal Pathophysiology, National Institute of Cardiology “Ignacio Chávez”, Mexico City 14080, Mexico; (O.E.A.-T.); (P.R.-M.); (L.G.S.-L.)
- Correspondence:
| |
Collapse
|
25
|
Faivre A, Scholz CC, de Seigneux S. Hypoxia in chronic kidney disease: towards a paradigm shift? Nephrol Dial Transplant 2020; 36:1782-1790. [PMID: 33895835 DOI: 10.1093/ndt/gfaa091] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Indexed: 11/15/2022] Open
Abstract
Chronic kidney disease (CKD) is defined as an alteration of kidney structure and/or function lasting for >3 months [1]. CKD affects 10% of the general adult population and is responsible for large healthcare costs [2]. Since the end of the last century, the role of hypoxia in CKD progression has controversially been discussed. To date, there is evidence of the presence of hypoxia in late-stage renal disease, but we lack time-course evidence, stage correlation and also spatial co-localization with fibrotic lesions to ensure its causative role. The classical view of hypoxia in CKD progression is that it is caused by peritubular capillary alterations, renal anaemia and increased oxygen consumption regardless of the primary injury. In this classical view, hypoxia is assumed to further induce pro-fibrotic and pro-inflammatory responses, as well as oxidative stress, leading to CKD worsening as part of a vicious circle. However, recent investigations tend to question this paradigm, and both the presence of hypoxia and its role in CKD progression are still not clearly demonstrated. Hypoxia-inducible factor (HIF) is the main transcriptional regulator of the hypoxia response. Genetic HIF modulation leads to variable effects on CKD progression in different murine models. In contrast, pharmacological modulation of the HIF pathway [i.e. by HIF hydroxylase inhibitors (HIs)] appears to be generally protective against fibrosis progression experimentally. We here review the existing literature on the role of hypoxia, the HIF pathway and HIF HIs in CKD progression and summarize the evidence that supports or rejects the hypoxia hypothesis, respectively.
Collapse
Affiliation(s)
- Anna Faivre
- Department of Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Carsten C Scholz
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,National Centre of Competence in Research "Kidney.CH", Zurich, Switzerland
| | - Sophie de Seigneux
- Department of Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland.,National Centre of Competence in Research "Kidney.CH", Zurich, Switzerland.,Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
26
|
Srivastava A, Prasad PV. Visualizing mitochondrial (dys)function using positron emission tomography imaging. Kidney Int 2020; 98:51-53. [PMID: 32571489 PMCID: PMC7745541 DOI: 10.1016/j.kint.2020.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 01/14/2023]
Abstract
Mitochondrial dysfunction is thought to be a critical pathway in the development and progression of kidney diseases, but optimal methods to assess kidney mitochondrial dysfunction are not well known. Saeki and colleagues use positron emission tomography imaging with a novel probe, 2-tert-butyl-4-chloro-5-[6-(4-18F-fluorobutoxy)-pyridin-3-ylmethoxy]-2H-pyridazin-3-one (18F-BCPP-BF), to visualize and assess kidney mitochondrial status. The authors demonstrate that reduced uptake of 18F-BCPP-BF, as assessed by positron emission tomography imaging, corresponds to reduced functioning mitochondria in 3 separate animal models of kidney diseases.
Collapse
Affiliation(s)
- Anand Srivastava
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Pottumarthi V Prasad
- Department of Radiology, NorthShore University HealthSystem, Evanston, Illinois, USA.
| |
Collapse
|
27
|
Saeki S, Ohba H, Ube Y, Tanaka K, Haruyama W, Uchii M, Kitayama T, Tsukada H, Shimada T. Positron emission tomography imaging of renal mitochondria is a powerful tool in the study of acute and progressive kidney disease models. Kidney Int 2020; 98:88-99. [DOI: 10.1016/j.kint.2020.02.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/06/2020] [Accepted: 02/20/2020] [Indexed: 01/28/2023]
|
28
|
Li Y, Nourbakhsh N, Pham H, Tham R, Zuckerman JE, Singh P. Evolution of altered tubular metabolism and mitochondrial function in sepsis-associated acute kidney injury. Am J Physiol Renal Physiol 2020; 319:F229-F244. [PMID: 32538150 DOI: 10.1152/ajprenal.00390.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sepsis-associated acute kidney injury (s-AKI) has a staggering impact in patients and lacks any treatment. Incomplete understanding of the pathogenesis of s-AKI is a major barrier to the development of effective therapies. We address the gaps in knowledge regarding renal oxygenation, tubular metabolism, and mitochondrial function in the pathogenesis of s-AKI using the cecal ligation and puncture (CLP) model in mice. At 24 h after CLP, renal oxygen delivery was reduced; however, fractional oxygen extraction was unchanged, suggesting inefficient renal oxygen utilization despite decreased glomerular filtration rate and filtered load. To investigate the underlying mechanisms, we examined temporal changes in mitochondrial function and metabolism at 4 and 24 h after CLP. At 4 h after CLP, markers of mitochondrial content and biogenesis were increased in CLP kidneys, but mitochondrial oxygen consumption rates were suppressed in proximal tubules. Interestingly, at 24 h, proximal tubular mitochondria displayed high respiratory capacity, but with decreased mitochondrial content, biogenesis, fusion, and ATP levels in CLP kidneys, suggesting decreased ATP synthesis efficiency. We further investigated metabolic reprogramming after CLP and observed reduced expression of fatty acid oxidation enzymes but increased expression of glycolytic enzymes at 24 h. However, assessment of functional glycolysis revealed lower glycolytic capacity, glycolytic reserve, and compensatory glycolysis in CLP proximal tubules, which may explain their susceptibility to injury. In conclusion, we demonstrated significant alterations in renal oxygenation, tubular mitochondrial function, and metabolic reprogramming in s-AKI, which may play an important role in the progression of injury and recovery from AKI in sepsis.
Collapse
Affiliation(s)
- Ying Li
- Division of Nephrology-Hypertension, University of California, San Diego, California.,Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Noureddin Nourbakhsh
- Division of Nephrology-Hypertension, University of California, San Diego, California.,Division of Pediatric Nephrology, Rady Children's Hospital San Diego, University of California, San Diego, California
| | - Hai Pham
- Division of Nephrology-Hypertension, University of California, San Diego, California.,Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Rick Tham
- Division of Nephrology-Hypertension, University of California, San Diego, California.,Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Jonathan E Zuckerman
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Prabhleen Singh
- Division of Nephrology-Hypertension, University of California, San Diego, California.,Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
29
|
Bush KT, Singh P, Nigam SK. Gut-derived uremic toxin handling in vivo requires OAT-mediated tubular secretion in chronic kidney disease. JCI Insight 2020; 5:133817. [PMID: 32271169 DOI: 10.1172/jci.insight.133817] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
The role of the renal organic anion transporters OAT1 (also known as SLC22A6, originally identified as NKT) and OAT3 (also known as SLC22A8) in chronic kidney disease (CKD) remains poorly understood. This is particularly so from the viewpoint of residual proximal tubular secretion, a key adaptive mechanism to deal with protein-bound uremic toxins in CKD. Using the subtotal nephrectomy (STN) model, plasma metabolites accumulating in STN rats treated with and without the OAT inhibitor, probenecid, were identified. Comparisons with metabolomics data from Oat1-KO and Oat3-KO mice support the centrality of the OATs in residual tubular secretion of uremic solutes, such as indoxyl sulfate, kynurenate, and anthranilate. Overlapping our data with those of published metabolomics data regarding gut microbiome-derived uremic solutes - which can have dual roles in signaling and toxicity - indicates that OATs play a critical role in determining their plasma levels in CKD. Thus, the OATs, along with other SLC and ABC drug transporters, are critical to the movement of uremic solutes across tissues and into various body fluids, consistent with the remote sensing and signaling theory. The data support a role for OATs in modulating remote interorganismal and interorgan communication (gut microbiota-blood-liver-kidney-urine). The results also have implications for understanding drug-metabolite interactions involving uremic toxins.
Collapse
Affiliation(s)
- Kevin T Bush
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Prabhleen Singh
- Division of Nephrology-Hypertension, University of California, San Diego and Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Sanjay K Nigam
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
30
|
The Urinary Exosomal miRNA Expression Profile is Predictive of Clinical Response in Lupus Nephritis. Int J Mol Sci 2020; 21:ijms21041372. [PMID: 32085620 PMCID: PMC7073236 DOI: 10.3390/ijms21041372] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 11/23/2022] Open
Abstract
Data on exosomal-derived urinary miRNAs have identified several miRNAs associated with disease activity and fibrosis formation, but studies on prognosis are lacking. We conducted a qPCR array screening on urinary exosomes from 14 patients with biopsy-proven proliferative lupus glomerulonephritis with a renal outcome of clinical response (n = 7) and non-response (n = 7) following therapy. Validation studies were performed by qRT-PCR in a new lupus nephritis (LN) cohort (responders = 22 and non-responders = 21). Responder patients expressed significantly increased levels of miR-31, miR-107, and miR-135b-5p in urine and renal tissue compared to non-responders. MiR-135b exhibited the best predictive value to discriminate responder patients (area under the curve = 0.783). In vitro studies showed exosome-derived miR-31, miR-107, and miR-135b-5p expression to be mainly produced by tubular renal cells stimulated with inflammatory cytokines (e.g IL1, TNFα, IFNα and IL6). Uptake of urinary exosomes from responders by mesangial cells was superior compared to that from non-responders (90% vs. 50%, p < 0.0001). HIF1A was identified as a potential common target, and low protein levels were found in non-responder renal biopsies. HIF1A inhibition reduced mesangial proliferation and IL-8, CCL2, CCL3, and CXCL1 mesangial cell production and IL-6/VCAM-1 in endothelial cells. Urinary exosomal miR-135b-5p, miR-107, and miR-31 are promising novel markers for clinical outcomes, regulating LN renal recovery by HIF1A inhibition.
Collapse
|
31
|
Abstract
Arterial hypertension has a large prevalence in the general population and as a major hypertensive target organ, the involvement of kidney is usually hard to avoid and gradually develops into chronic kidney disease (CKD). Acute hypertension is defined as a blood pressure greater than 180/120, also known as hypertensive emergency (HE). In acute severe hypertension, the pathophysiology damage to the kidney tends to worsen on the basis of chronic damage, and accounts for more significant mortality. However, the mechanisms of renal injury induced by acute hypertension remain unclear. This review summarizes the clinical and histopathological features of hypertensive renal injury by using "in vivo cyrotechnique" and focusses on the interplay of distinct systemic signaling pathways, which drive glomerular podocyte injury. A thorough understanding of the cellular and molecular mechanisms of kidney damage and repair in hypertension will provide significant insight into the development of new research methods and therapeutic strategies for global CKD progression.
Collapse
|
32
|
Molecular Mechanisms of Kidney Injury and Repair in Arterial Hypertension. Int J Mol Sci 2019; 20:ijms20092138. [PMID: 31052201 PMCID: PMC6539752 DOI: 10.3390/ijms20092138] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/25/2019] [Accepted: 04/28/2019] [Indexed: 02/06/2023] Open
Abstract
The global burden of chronic kidney disease is rising. The etiologies, heterogeneous, and arterial hypertension, are key factors contributing to the development and progression of chronic kidney disease. Arterial hypertension is induced and maintained by a complex network of systemic signaling pathways, such as the hormonal axis of the renin-angiotensin-aldosterone system, hemodynamic alterations affecting blood flow, oxygen supply, and the immune system. This review summarizes the clinical and histopathological features of hypertensive kidney injury and focusses on the interplay of distinct systemic signaling pathways, which drive hypertensive kidney injury in distinct cell types of the kidney. There are several parallels between hypertension-induced molecular signaling cascades in the renal epithelial, endothelial, interstitial, and immune cells. Angiotensin II signaling via the AT1R, hypoxia induced HIFα activation and mechanotransduction are closely interacting and further triggering the adaptions of metabolism, cytoskeletal rearrangement, and profibrotic TGF signaling. The interplay of these, and other cellular pathways, is crucial to balancing the injury and repair of the kidneys and determines the progression of hypertensive kidney disease.
Collapse
|
33
|
Cippà PE, Sun B, Liu J, Chen L, Naesens M, McMahon AP. Transcriptional trajectories of human kidney injury progression. JCI Insight 2018; 3:123151. [PMID: 30429361 DOI: 10.1172/jci.insight.123151] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/10/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The molecular understanding of the progression from acute to chronic organ injury is limited. Ischemia/reperfusion injury (IRI) triggered during kidney transplantation can contribute to progressive allograft dysfunction. METHODS Protocol biopsies (n = 163) were obtained from 42 kidney allografts at 4 time points after transplantation. RNA sequencing-mediated (RNA-seq-mediated) transcriptional profiling and machine learning computational approaches were employed to analyze the molecular responses to IRI and to identify shared and divergent transcriptional trajectories associated with distinct clinical outcomes. The data were compared with the response to IRI in a mouse model of the acute to chronic kidney injury transition. RESULTS In the first hours after reperfusion, all patients exhibited a similar transcriptional program under the control of immediate-early response genes. In the following months, we identified 2 main transcriptional trajectories leading to kidney recovery or to sustained injury with associated fibrosis and renal dysfunction. The molecular map generated by this computational approach highlighted early markers of kidney disease progression and delineated transcriptional programs associated with the transition to chronic injury. The characterization of a similar process in a mouse IRI model extended the relevance of our findings beyond transplantation. CONCLUSIONS The integration of multiple transcriptomes from serial biopsies with advanced computational algorithms overcame the analytical hurdles related to variability between individuals and identified shared transcriptional elements of kidney disease progression in humans, which may prove as useful predictors of disease progression following kidney transplantation and kidney injury. This generally applicable approach opens the way for an unbiased analysis of human disease progression. FUNDING The study was supported by the California Institute for Regenerative Medicine and by the Swiss National Science Foundation.
Collapse
Affiliation(s)
- Pietro E Cippà
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California (USC), Los Angeles, USA.,Division of Nephrology, Regional Hospital Lugano, Lugano, Switzerland
| | - Bo Sun
- Molecular and Computational Biology, USC, Los Angeles, USA
| | - Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California (USC), Los Angeles, USA
| | - Liang Chen
- Molecular and Computational Biology, USC, Los Angeles, USA
| | - Maarten Naesens
- Department of Microbiology and Immunology, KU Leuven, and Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California (USC), Los Angeles, USA
| |
Collapse
|
34
|
Mafra D, Gidlund EK, Borges NA, Magliano DC, Lindholm B, Stenvinkel P, von Walden F. Bioactive food and exercise in chronic kidney disease: Targeting the mitochondria. Eur J Clin Invest 2018; 48:e13020. [PMID: 30144313 DOI: 10.1111/eci.13020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 08/11/2018] [Accepted: 08/22/2018] [Indexed: 12/16/2022]
Abstract
Chronic kidney disease (CKD), which affects 10%-15% of the population, associates with a range of complications-such as cardiovascular disease, frailty, infections, muscle and bone disorders and premature ageing-that could be related to alterations of mitochondrial number, distribution, structure and function. As mitochondrial biogenesis, bioenergetics and the dynamic mitochondrial networks directly or indirectly regulate numerous intra- and extracellular functions, the mitochondria have emerged as an important target for interventions aiming at preventing or improving the treatment of complications in CKD. In this review, we discuss the possible role of bioactive food compounds and exercise in the modulation of the disturbed mitochondrial function in a uraemic milieu.
Collapse
Affiliation(s)
- Denise Mafra
- Graduate Program in Medical Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil.,Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Eva-Karin Gidlund
- Division of Molecular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Natália Alvarenga Borges
- Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - D'Angelo Carlo Magliano
- Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Bengt Lindholm
- Division of Renal Medicine, Department of Clinical Science Intervention and Technology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science Intervention and Technology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ferdinand von Walden
- Division of Pediatric Neurology, Department of Women's and Children's health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
Mitochondrial bioenergetics, redox state, dynamics and turnover alterations in renal mass reduction models of chronic kidney diseases and their possible implications in the progression of this illness. Pharmacol Res 2018; 135:1-11. [PMID: 30030169 DOI: 10.1016/j.phrs.2018.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 01/13/2023]
Abstract
Nowadays, chronic kidney disease (CKD) is considered a worldwide public health problem. CKD is a term used to describe a set of pathologies that structurally and functionally affect the kidney, it is mostly characterized by the progressive loss of kidney function. Current therapeutic approaches are insufficient to avoid the development of this disease, which highlights the necessity of developing new strategies to reverse or at least delay CKD progression. Kidney is highly dependent on mitochondrial homeostasis and function, consequently, the idea that mitochondrial pathologies could play a pivotal role in the genesis and development of kidney diseases has risen. Although many research groups have recently published studies of mitochondrial function in acute kidney disease models, the existing information about CKD is still limited, especially in renal mass reduction (RMR) models. This paper focuses on reviewing current experimental information about the bioenergetics, dynamics (fission and fusion processes), turnover (mitophagy and biogenesis) and redox mitochondrial alterations in RMR, to discuss and integrate the mitochondrial changes triggered by nephron loss, as well as its relationship with loss of kidney function in CKD, in these models. Understanding these mechanisms would allow us to design new therapies that target these mitochondrial alterations.
Collapse
|
36
|
Mitochondrial Dysfunction and Signaling in Diabetic Kidney Disease: Oxidative Stress and Beyond. Semin Nephrol 2018; 38:101-110. [DOI: 10.1016/j.semnephrol.2018.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
37
|
Sun L, Yuan Q, Xu T, Yao L, Feng J, Ma J, Wang L, Lu C, Wang D. Pioglitazone Improves Mitochondrial Function in the Remnant Kidney and Protects against Renal Fibrosis in 5/6 Nephrectomized Rats. Front Pharmacol 2017; 8:545. [PMID: 28860994 PMCID: PMC5559534 DOI: 10.3389/fphar.2017.00545] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/04/2017] [Indexed: 11/13/2022] Open
Abstract
Pioglitazone is a type of peroxisome proliferator-activated receptor γ (PPARγ) agonist and has been demonstrated to be effective in chronic kidney diseases (CKD) treatment. However, the underlying mechanism involved in the renoprotection of pioglitazone has not been fully revealed. In the present study, the renoprotective mechanism of pioglitazone was investigated in 5/6 nephrectomized (Nx) rats and TGF-β1-exposed HK-2 cells. Pioglitazone attenuated renal injury and improved renal function, as examined by 24 h urinary protein, blood urea nitrogen and plasma creatinine in Nx rats. Renal fibrosis and enhanced expressions of profibrotic proteins TGF-β1, fibronectin and collagen I caused by Nx were significantly alleviated by pioglitazone. In addition, pioglitazone protected mitochondrial functions by stabilizing the mitochondrial membrane potential, inhibiting ROS generation, maintaining ATP production and the activities of complexes I and III, and preventing cytochrome C leakage from mitochondria. Pioglitazone also upregulated the expression levels of ATP synthase β, COX I and NDUFB8, which were downregulated in the kidney of Nx rats and TGF-β1-exposed HK-2 cells. Furthermore, pioglitazone increased fusion proteins Opa-1 and Mfn2 expressions and decreased fission protein Drp1 expression. The results imply that pioglitazone may exert the renoprotective effects through modulating mitochondrial electron transport chain and mitochondrial dynamics in CKD. Finally, these recoveries were completely or partly inhibited by GW9662, which suggests that these effects at least partly PPARγ dependent. This study provides evidence for the pharmacological mechanism of pioglitazone in the treatment of CKD.
Collapse
Affiliation(s)
- Li Sun
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang, China
| | - Quan Yuan
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Tianhua Xu
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang, China
| | - Li Yao
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang, China
| | - Jiangmin Feng
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang, China
| | - Jianfei Ma
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang, China
| | - Lining Wang
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang, China
| | - Changlong Lu
- Department of Immunology, China Medical UniversityShenyang, China
| | - Danan Wang
- Department of Immunology, China Medical UniversityShenyang, China
| |
Collapse
|