1
|
Wang Y, Ding Y, Dong H, Wuren T, Luo P. MSCs in Acute Kidney Injury Treatment: Modulating Mitochondrial Function and Inhibiting Pyroptosis via PGC-1α. Exp Cell Res 2025:114583. [PMID: 40324626 DOI: 10.1016/j.yexcr.2025.114583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE This study aims to investigate the mechanisms of MSC therapy for acute kidney injury, focusing on the regulation of mitochondrial function and pyroptosis in renal tubular epithelial cells (RTECs). METHODS An in vivo ischemia/reperfusion (I/R) model was used to assess the effects of MSC treatment on mitochondrial membrane potential, mitochondrial function, cell pyroptosis, and PGC-1α expression in RTECs. RESULTS MSCs significantly improved mitochondrial function in RTECs by upregulating PGC-1α expression, regulating mitochondrial fusion and fission proteins, reducing mitochondrial ROS production, and suppressing NLRP3 inflammasome activation. Furthermore, MSC treatment reduced the levels of pyroptotic markers, such as IL-18, and exhibited a marked anti-fibrotic effect in the long-term. These findings suggest that MSCs not only repair acute kidney injury but also offer long-term protection against fibrosis. CONCLUSION MSCs improve the repair of acute kidney injury by modulating mitochondrial function and inhibiting pyroptosis, providing new theoretical support for MSC-based therapies in AKI treatment.
Collapse
Affiliation(s)
- Yanjun Wang
- Department of Nephrology, Affiliated Hospital of Qinghai University, Xining, Qinghai 810001,China; Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810016,China; High-Altitude Medicine Key Laboratory of the Ministry of Educationy, Xining, Qinghai 810001,China; Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine (Qinghai-Utah Joint Key Laboratory for Plateau Medicine), Xining, Qinghai 810001,China
| | - Yanlin Ding
- Department of Nephrology, Affiliated Hospital of Qinghai University, Xining, Qinghai 810001,China
| | - Haiyun Dong
- Department of Nephrology, Affiliated Hospital of Qinghai University, Xining, Qinghai 810001,China
| | - Tana Wuren
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810016,China; High-Altitude Medicine Key Laboratory of the Ministry of Educationy, Xining, Qinghai 810001,China; Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine (Qinghai-Utah Joint Key Laboratory for Plateau Medicine), Xining, Qinghai 810001,China
| | - Pengli Luo
- Department of Nephrology, Affiliated Hospital of Qinghai University, Xining, Qinghai 810001,China.
| |
Collapse
|
2
|
Tiscornia C, Tapia V, Águila D, Lorca-Ponce E, Aicardi V, Vásquez F. Maqui and Chronic Kidney Disease: A Narrative Review on the Potential Nephroprotective Role of Anthocyanins. Nutrients 2025; 17:1058. [PMID: 40292440 PMCID: PMC11944665 DOI: 10.3390/nu17061058] [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: 01/29/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Chronic kidney disease (CKD) is a progressive pathology, with high global prevalence, associated with inflammation and oxidative stress. Given the limited capacity of conventional treatments to reverse renal damage, complementary alternatives have emerged such as supplementation with anthocyanins from maqui (Aristotelia chilensis), known for their antioxidant and anti-inflammatory properties. This review analyzes the evidence for their impact on CKD progression. Methods: A narrative review of the experimental literature regarding maqui anthocyanins, their bioavailability, and their effects on oxidative stress, inflammation, and CKD to January 2025 was conducted. Articles without peer review or without a focus on Aristotelia chilensis were excluded, guaranteeing an updated compilation on its nephroprotective potential. Results: Anthocyanins have shown benefits in reducing oxidative stress, inflammation, and glycemia regulation. Preclinical studies suggest improvements in renal function as well as less fibrosis. Human trials indicate positive effects on metabolism, although evidence in CKD patients is limited. Bioavailability remains a challenge to optimizing efficacy. Conclusions: Maqui is a promising source of anthocyanins, with nephroprotective potential. However, robust clinical studies are required to determine its safety, optimal dose, and long-term impact in CKD. Its incorporation into evidence-based therapeutic strategies could offer an innovative approach in the management of this disease. More clinical studies are needed to validate the preclinical findings and optimize the therapeutic use of maqui in CKD.
Collapse
Affiliation(s)
- Caterina Tiscornia
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501014, Chile; (C.T.); (V.T.); (D.Á.)
| | - Violeta Tapia
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501014, Chile; (C.T.); (V.T.); (D.Á.)
| | - Daniela Águila
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501014, Chile; (C.T.); (V.T.); (D.Á.)
| | - Enrique Lorca-Ponce
- Escuela de Enfermería, Universidad Finis Terrae, Santiago 7501014, Chile;
- Escuela de Kinesiología, Facultad de Arte y Educación Física, Universidad Metropolitana en Ciencias de la Santiago, Santiago 7760197, Chile
| | - Valeria Aicardi
- Unidad de Diálisis, Clínica Indisa, Santiago 7501014, Chile;
| | - Fabián Vásquez
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501014, Chile; (C.T.); (V.T.); (D.Á.)
| |
Collapse
|
3
|
Yao C, Li Z, Sun K, Zhang Y, Shou S, Jin H. Mitochondrial dysfunction in acute kidney injury. Ren Fail 2024; 46:2393262. [PMID: 39192578 PMCID: PMC11360640 DOI: 10.1080/0886022x.2024.2393262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Acute kidney injury (AKI) is a systemic clinical syndrome increasing morbidity and mortality worldwide in recent years. Renal tubular epithelial cells (TECs) death caused by mitochondrial dysfunction is one of the pathogeneses. The imbalance of mitochondrial quality control is the main cause of mitochondrial dysfunction. Mitochondrial quality control plays a crucial role in AKI. Mitochondrial quality control mechanisms are involved in regulating mitochondrial integrity and function, including antioxidant defense, mitochondrial quality control, mitochondrial DNA (mtDNA) repair, mitochondrial dynamics, mitophagy, and mitochondrial biogenesis. Currently, many studies have used mitochondrial dysfunction as a targeted therapeutic strategy for AKI. Therefore, this review aims to present the latest research advancements on mitochondrial dysfunction in AKI, providing a valuable reference and theoretical foundation for clinical prevention and treatment of this condition, ultimately enhancing patient prognosis.
Collapse
Affiliation(s)
- Congcong Yao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Ziwei Li
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Keke Sun
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Zhang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Songtao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Heng Jin
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Choi DH, Lee SM, Park BN, Lee MH, Yang DE, Son YK, Kim SE, An WS. Omega-3 Fatty Acids Modify Drp1 Expression and Activate the PINK1-Dependent Mitophagy Pathway in the Kidney and Heart of Adenine-Induced Uremic Rats. Biomedicines 2024; 12:2107. [PMID: 39335620 PMCID: PMC11429207 DOI: 10.3390/biomedicines12092107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Mitochondrial homeostasis is controlled by biogenesis, dynamics, and mitophagy. Mitochondrial dysfunction plays a central role in cardiovascular and renal disease and omega-3 fatty acids (FAs) are beneficial for cardiovascular disease. We investigated whether omega-3 fatty acids (FAs) regulate mitochondrial biogenesis, dynamics, and mitophagy in the kidney and heart of adenine-induced uremic rats. Eighteen male Sprague Dawley rats were divided into normal control, adenine control, and adenine with omega-3 FA groups. Using Western blot analysis, the kidney and heart expression of mitochondrial homeostasis-related molecules, including peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), dynamin-related protein 1 (Drp1), and phosphatase and tensin homolog-induced putative kinase 1 (PINK1) were investigated. Compared to normal, serum creatinine and heart weight/body weight in adenine control were increased and slightly improved in the omega-3 FA group. Compared to the normal controls, the expression of PGC-1α and PINK1 in the kidney and heart of the adenine group was downregulated, which was reversed after omega-3 FA supplementation. Drp1 was upregulated in the kidney but downregulated in the heart in the adenine group. Drp1 expression in the heart recovered in the omega-3 FA group. Mitochondrial DNA (mtDNA) was decreased in the kidney and heart of the adenine control group but the mtDNA of the heart was recovered in the omega-3 FA group. Drp1, which is related to mitochondrial fission, may function oppositely in the uremic kidney and heart. Omega-3 FAs may be beneficial for mitochondrial homeostasis by activating mitochondrial biogenesis and PINK1-dependent mitophagy in the kidney and heart of uremic rats.
Collapse
Affiliation(s)
- Dong Ho Choi
- Department of Internal Medicine, Good Moon Hwa Hospital, Busan 48735, Republic of Korea
| | - Su Mi Lee
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Bin Na Park
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Mi Hwa Lee
- Department of Anatomy and Cell Biology, Dong-A University, Busan 49201, Republic of Korea;
| | - Dong Eun Yang
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Young Ki Son
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Seong Eun Kim
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Won Suk An
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
- Medical Science Research Center, Dong-A University, Busan 49201, Republic of Korea
| |
Collapse
|
5
|
Zhu M, Zeiss C, Hamrick MW, Weinstein RS, Sun BH, Brotto M, Liu X, Siu E, Huttner A, Tommasini S, Simpson C, Insogna K. Mitofusin 2 plays a critical role in maintaining the functional integrity of the neuromuscular-skeletal axis. Bone 2024; 184:117086. [PMID: 38552893 DOI: 10.1016/j.bone.2024.117086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE Mitofusin 2 (Mfn2) is one of two mitofusins involved in regulating mitochondrial size, shape and function, including mitophagy, an important cellular mechanism to limit oxidative stress. Reduced expression of Mfn2 has been associated with impaired osteoblast differentiation and function and a reduction in the number of viable osteocytes in bone. We hypothesized that the genetic absence of Mfn2 in these cells would increase their susceptibility to aging-associated metabolic stress, leading to a progressive impairment in skeletal homeostasis over time. METHODS Mfn2 was selectively deleted in vivo at three different stages of osteoblast lineage commitment by crossing mice in which the Mfn2 gene was floxed with transgenic mice expressing Cre under the control of the promoter for Osterix (OSX), collagen1a1, or DMP1 (Dentin Matrix Acidic Phosphoprotein 1). RESULTS Mice in which Mfn2 was deleted using DMP1-cre demonstrated a progressive and dramatic decline in bone mineral density (BMD) beginning at 10 weeks of age (n = 5 for each sex and each genotype from age 10 to 20 weeks). By 15 weeks, there was evidence for a functional decline in muscle performance as assessed using a rotarod apparatus (n = 3; 2 males/ 1 female for each genotype), accompanied by a decline in lean body mass. A marked reduction in trabecular bone mass was evident on bone histomorphometry, and biomechanical testing at 25 weeks (k/o: 2 male/1 female, control 2 male/2 female) revealed severely impaired femur strength. Extensive regional myofiber atrophy and degeneration was observed on skeletal muscle histology. Electron microscopy showed progressive disruption of cellular architecture, with disorganized sarcomeres and a bloated mitochondrial reticulum. There was also evidence of neurodegeneration within the ventral horn and roots of the lumbar spinal cord, which was accompanied by myelin loss and myofiber atrophy. Deletion of Mfn2 using OSX-cre or Col1a1-cre did not result in a musculoskeletal phenotype. Where possible, male and female animals were analyzed separately, but small numbers of animals in each group limited statistical power. For other outcomes, where sex was not considered, small sample sizes might still limit the strength of the observation. CONCLUSION Despite known functional overlap of Mfn1 and Mfn2 in some tissues, and their co-expression in bone, muscle and spinal cord, deletion of Mfn2 using the 8 kB DMP1 promoter uncovered an important non-redundant role for Mfn2 in maintaining the neuromuscular/bone axis.
Collapse
Affiliation(s)
- Meiling Zhu
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA
| | - Caroline Zeiss
- Yale School of Medicine, Section of Comparative Medicine, New Haven, CT, USA
| | - Mark W Hamrick
- Department of Orthopaedic Surgery, Institute of Molecular Medicine & Genetics, Medical College of Georgia, Augusta, GA, USA
| | - Robert S Weinstein
- Division of Endocrinology, Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ben-Hua Sun
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX, USA
| | - Xinran Liu
- Yale School of Medicine, Center for Cellular and Molecular Imaging, New Haven, CT, USA
| | - Edwin Siu
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA
| | - Anita Huttner
- Yale School of Medicine, Department of Pathology, New Haven, CT, USA
| | - Steven Tommasini
- Yale School of Medicine, Department of Orthopaedics & Rehabilitation, New Haven, CT, USA
| | - Christine Simpson
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA
| | - Karl Insogna
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA.
| |
Collapse
|
6
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
7
|
Chien MJ, Li SJ, Wong SC, Chiang CH, Lin YY, Mersmann HJ, Chen CY. Determination of mitochondrial functions and damage in kidney in female LeeSung minipigs with a high-fat diet-induced obesity. Arch Physiol Biochem 2023; 129:1289-1297. [PMID: 34338085 DOI: 10.1080/13813455.2021.1949022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/23/2021] [Indexed: 10/20/2022]
Abstract
The purpose of this study was to investigate the nexus between mitochondrial function and kidney injury by using a dietary-induced obese minipig model. Female Lee-Sung minipigs feeding a high-fat diet (HFD) for 6 months exhibited obesity, hyperglycaemia and dyslipidemia. HFD elevated the levels of plasma biomarkers related to renal injury, including symmetric dimethylarginine, creatinine and urea nitrogen. An extensive structural change in tubules and glomeruli was observed in HFD-fed pigs. A great amount of triacylglycerol was accumulated in HFD kidney compared to control kidney, whereas a reduction of ATP level and antioxidant capacity were exhibited in HFD kidney. Moreover, HFD altered the expressions of mitochondrial-related protein in renal cortex. To conclude, long-term HFD feeding to Lee-Sung minipigs induced obesity and kidney injury accompanied by abnormal mitochondrial functions in the renal cortex, suggesting an interrelationship with renal disease progression.
Collapse
Affiliation(s)
- Miao-Ju Chien
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Sin-Jin Li
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Shiu-Chung Wong
- National Taiwan University Veterinary Hospital, National Taiwan University, Taipei, Taiwan
| | - Chun-Hsien Chiang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yuan-Yu Lin
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Harry J Mersmann
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Ching-Yi Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
8
|
Wang S, Sang X, Li S, Yang W, Wang S, Chen H, Lu C. Increased Ca2 + transport across the mitochondria-associated membranes by Mfn2 inhibiting endoplasmic reticulum stress in ischemia/reperfusion kidney injury. Sci Rep 2023; 13:17257. [PMID: 37828353 PMCID: PMC10570331 DOI: 10.1038/s41598-023-44538-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023] Open
Abstract
Renal ischemia/reperfusion (I/R) injury, which leads to acute kidney injury (AKI), is a major cause of morbidity and mortality in a variety of clinical situations. This study aimed to investigate the protective role of Mfn2 during renal I/R injury. Overexpression of Mfn2 in NRK-52E rat renal tubular epithelial cells and rats, then we constructed hypoxia reoxygenation (H/R) cells and I/R rat model. Apoptosis, ROS, ATP, Ca2+ levels in cells and rats, as well as renal tissue and functional injury in rats were detected respectively. Endoplasmic reticulum (ER) stress was further examined in cells and rats. The morphological changes of mitochondria-associated ER membranes (MAMs) were also detected. Mfn2 expression is reduced in H/R-treated NRK-52E cells and renal tissue of I/R rats. At the cellular level, overexpression of Mfn2 promoted cell proliferation, inhibited cell apoptosis, attenuated mitochondrial damage and Ca2+ overload, and ER stress. In addition, Mfn2 also restored the MAMs structure. In vivo experiments found that overexpression of Mfn2 could improve renal function and alleviate tissue injury. Concomitant with elevated Mfn2 expression in the kidney, reduced renal cell apoptosis, restored mitochondrial function, and reduced calcium overload. Finally, ER stress in rat kidney tissue was alleviated after overexpression of Mfn2. These results reveal that Mfn2 contributes to ER stress, mitochondrial function, and cell death in I/R injury, which provides a novel therapeutic target for AKI.
Collapse
Affiliation(s)
- Shun Wang
- Nephrology Center, The First Affiliated Hospital of Xinjiang Medical University, Xinshi District, Urumqi, 830054, China
| | - Xiaohong Sang
- Nephrology Center, The First Affiliated Hospital of Xinjiang Medical University, Xinshi District, Urumqi, 830054, China
| | - Suhua Li
- Nephrology Center, The First Affiliated Hospital of Xinjiang Medical University, Xinshi District, Urumqi, 830054, China
| | - Wenjun Yang
- Nephrology Center, The First Affiliated Hospital of Xinjiang Medical University, Xinshi District, Urumqi, 830054, China
| | - Shihan Wang
- Nephrology Center, The First Affiliated Hospital of Xinjiang Medical University, Xinshi District, Urumqi, 830054, China
| | - Haixia Chen
- Nephrology Center, The First Affiliated Hospital of Xinjiang Medical University, Xinshi District, Urumqi, 830054, China
| | - Chen Lu
- Nephrology Center, The First Affiliated Hospital of Xinjiang Medical University, Xinshi District, Urumqi, 830054, China.
| |
Collapse
|
9
|
Bhatia D, Choi ME. Autophagy and mitophagy: physiological implications in kidney inflammation and diseases. Am J Physiol Renal Physiol 2023; 325:F1-F21. [PMID: 37167272 PMCID: PMC10292977 DOI: 10.1152/ajprenal.00012.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023] Open
Abstract
Autophagy is a ubiquitous intracellular cytoprotective quality control program that maintains cellular homeostasis by recycling superfluous cytoplasmic components (lipid droplets, protein, or glycogen aggregates) and invading pathogens. Mitophagy is a selective form of autophagy that by recycling damaged mitochondrial material, which can extracellularly act as damage-associated molecular patterns, prevents their release. Autophagy and mitophagy are indispensable for the maintenance of kidney homeostasis and exert crucial functions during both physiological and disease conditions. Impaired autophagy and mitophagy can negatively impact the pathophysiological state and promote its progression. Autophagy helps in maintaining structural integrity of the kidney. Mitophagy-mediated mitochondrial quality control is explicitly critical for regulating cellular homeostasis in the kidney. Both autophagy and mitophagy attenuate inflammatory responses in the kidney. An accumulating body of evidence highlights that persistent kidney injury-induced oxidative stress can contribute to dysregulated autophagic and mitophagic responses and cell death. Autophagy and mitophagy also communicate with programmed cell death pathways (apoptosis and necroptosis) and play important roles in cell survival by preventing nutrient deprivation and regulating oxidative stress. Autophagy and mitophagy are activated in the kidney after acute injury. However, their aberrant hyperactivation can be deleterious and cause tissue damage. The findings on the functions of autophagy and mitophagy in various models of chronic kidney disease are heterogeneous and cell type- and context-specific dependent. In this review, we discuss the roles of autophagy and mitophagy in the kidney in regulating inflammatory responses and during various pathological manifestations.
Collapse
Affiliation(s)
- Divya Bhatia
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, United States
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, United States
| |
Collapse
|
10
|
Barbero NM, Oller J, Sanz AB, Ramos AM, Ortiz A, Ruiz-Ortega M, Rayego-Mateos S. Mitochondrial Dysfunction in the Cardio-Renal Axis. Int J Mol Sci 2023; 24:ijms24098209. [PMID: 37175915 PMCID: PMC10179675 DOI: 10.3390/ijms24098209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Cardiovascular disease (CVD) frequently complicates chronic kidney disease (CKD). The risk of all-cause mortality increases from 20% to 500% in patients who suffer both conditions; this is referred to as the so-called cardio-renal syndrome (CRS). Preclinical studies have described the key role of mitochondrial dysfunction in cardiovascular and renal diseases, suggesting that maintaining mitochondrial homeostasis is a promising therapeutic strategy for CRS. In this review, we explore the malfunction of mitochondrial homeostasis (mitochondrial biogenesis, dynamics, oxidative stress, and mitophagy) and how it contributes to the development and progression of the main vascular pathologies that could be affected by kidney injury and vice versa, and how this knowledge may guide the development of novel therapeutic strategies in CRS.
Collapse
Affiliation(s)
- Nerea Mendez Barbero
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Jorge Oller
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Ana B Sanz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Adrian M Ramos
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Marta Ruiz-Ortega
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| | - Sandra Rayego-Mateos
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| |
Collapse
|
11
|
Chen RY, Li DW, Xie H, Liu XW, Zhuang SY, Wu HY, Wu JJ, Sun N, Qu JW, Miao JY, Zhong C, Huang YH, Yuan XD, Zhang M, Zhang WJ, Hou JQ. Gene signature and prediction model of the mitophagy-associated immune microenvironment in renal ischemia-reperfusion injury. Front Immunol 2023; 14:1117297. [PMID: 37056767 PMCID: PMC10086170 DOI: 10.3389/fimmu.2023.1117297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundRenal ischemia-reperfusion injury (IRI) is an inevitable occurrence during kidney transplantation. Mitophagy, ferroptosis, and the associated immune microenvironment (IME) have been shown to play important roles in renal IRI. However, the role of mitophagy-associated IME genes in IRI remains unclear. In this study, we aimed to construct a prediction model of IRI prognosis based on mitophagy-associated IME genes.MethodThe specific biological characteristics of the mitophagy-associated IME gene signature were comprehensively analyzed using public databases such as GEO, Pathway Unification, and FerrDb. Correlations between the expression of prognostic genes and immune-related genes and IRI prognosis were determined by Cox regression, LASSO analysis, and Pearson’s correlation. Molecular validation was performed using human kidney 2 (HK2) cells and culture supernatant as well as the serum and kidney tissues of mice after renal IRI. Gene expression was measured by PCR, and inflammatory cell infiltration was examined by ELISA and mass cytometry. Renal tissue damage was characterized using renal tissue homogenate and tissue sections.ResultsThe expression of the mitophagy-associated IME gene signature was significantly correlated with IRI prognosis. Excessive mitophagy and extensive immune infiltration were the primary factors affecting IRI. In particular, FUNDC1, SQSTM1, UBB, UBC, KLF2, CDKN1A, and GDF15 were the key influencing factors. In addition, B cells, neutrophils, T cells, and M1 macrophages were the key immune cells present in the IME after IRI. A prediction model for IRI prognosis was constructed based on the key factors associated with the mitophagy IME. Validation experiments in cells and mice indicated that the prediction model was reliable and applicable.ConclusionWe clarified the relationship between the mitophagy-related IME and IRI. The IRI prognostic prediction model based on the mitophagy-associated IME gene signature provides novel insights on the prognosis and treatment of renal IRI.
Collapse
Affiliation(s)
- Ruo-Yang Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Da-Wei Li
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Hui Xie
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Xiao-Wen Liu
- Department of Institute of Molecular Medicine, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Shao-Yong Zhuang
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Hao-Yu Wu
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jia-Jin Wu
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Nan Sun
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jun-Wen Qu
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jia-Yi Miao
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Chen Zhong
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Yu-Hua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Dong Yuan
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
- *Correspondence: Xiao-Dong Yuan, ; Ming Zhang, ; Wei-Jie Zhang, ; Jian-Quan Hou,
| | - Ming Zhang
- Department of Urology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
- *Correspondence: Xiao-Dong Yuan, ; Ming Zhang, ; Wei-Jie Zhang, ; Jian-Quan Hou,
| | - Wei-Jie Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Xiao-Dong Yuan, ; Ming Zhang, ; Wei-Jie Zhang, ; Jian-Quan Hou,
| | - Jian-Quan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
- *Correspondence: Xiao-Dong Yuan, ; Ming Zhang, ; Wei-Jie Zhang, ; Jian-Quan Hou,
| |
Collapse
|
12
|
Oxidative Stress and Mitochondrial Dysfunction in Chronic Kidney Disease. Cells 2022; 12:cells12010088. [PMID: 36611880 PMCID: PMC9818928 DOI: 10.3390/cells12010088] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The kidney contains many mitochondria that generate ATP to provide energy for cellular processes. Oxidative stress injury can be caused by impaired mitochondria with excessive levels of reactive oxygen species. Accumulating evidence has indicated a relationship between oxidative stress and kidney diseases, and revealed new insights into mitochondria-targeted therapeutics for renal injury. Improving mitochondrial homeostasis, increasing mitochondrial biogenesis, and balancing mitochondrial turnover has the potential to protect renal function against oxidative stress. Although there are some reviews that addressed this issue, the articles summarizing the relationship between mitochondria-targeted effects and the risk factors of renal failure are still few. In this review, we integrate recent studies on oxidative stress and mitochondrial function in kidney diseases, especially chronic kidney disease. We organized the causes and risk factors of oxidative stress in the kidneys based in their mitochondria-targeted effects. This review also listed the possible candidates for clinical therapeutics of kidney diseases by modulating mitochondrial function.
Collapse
|
13
|
Ramaiah P, Patra I, Abbas A, Fadhil AA, Abohassan M, Al-Qaim ZH, Hameed NM, Al-Gazally ME, Kemil Almotlaq SS, Mustafa YF, Shiravand Y. Mitofusin-2 in cancer: Friend or foe? Arch Biochem Biophys 2022; 730:109395. [PMID: 36176224 DOI: 10.1016/j.abb.2022.109395] [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: 07/02/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022]
Abstract
Cancer is a category of disorders characterized by excessive cell proliferation with the ability to infiltrate or disseminate to other organs of the body. Mitochondrial dysfunction, as one of the most prominent hallmarks of cancer cells, has been related to the onset and development of various cancers. Mitofusin 2 (MFN2) is a major mediator of mitochondrial fusion, endoplasmic reticulum (ER)-mitochondria interaction, mitophagy and axonal transport of mitochondria. Available data have shown that MFN2, which its alterations have been associated with mitochondrial dysfunction, could affect cancer initiation and progression. In fact, it showed that MFN2 may have a double-edged sword effect on cancer fate. Precisely, it demonstrated that MFN2, as a tumor suppressor, induces cancer cell apoptosis and inhibits cell proliferation via Ca2+ and Bax-mediated apoptosis and increases P21 and p27 levels, respectively. It also could suppress cell survival via inhibiting PI3K/Akt, Ras-ERK1/2-cyclin D1 and mTORC2/Akt signaling pathways. On the other hand, MFN2, as an oncogene, could increase cancer invasion via snail-mediated epithelial-mesenchymal transition (EMT) and in vivo tumorigenesis. While remarkable progress has been achieved in recent decades, further exploration is required to elucidate whether MFN2 could be a friend or it's an enemy. This study aimed to highlight the different functions of MFN2 in various cancers.
Collapse
Affiliation(s)
| | | | - Anum Abbas
- Basic Health Unit, Foundation University Medical College, Islamabad, Pakistan.
| | - Ali Abdulhussain Fadhil
- College of Medical Technology, Medical Lab Techniques, Al-farahidi University, Baghdad, Iraq
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 9088, Saudi Arabia
| | | | | | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul-41001, Iraq
| | - Yavar Shiravand
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80138, Naples, Italy.
| |
Collapse
|
14
|
Hepokoski M, Singh P. Mitochondria as mediators of systemic inflammation and organ cross talk in acute kidney injury. Am J Physiol Renal Physiol 2022; 322:F589-F596. [PMID: 35379000 PMCID: PMC9054254 DOI: 10.1152/ajprenal.00372.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acute kidney injury (AKI) is a systemic inflammatory disease that contributes to remote organ failures. Multiple organ failure is the leading cause of death due to AKI, and lack of understanding of the mechanisms involved has precluded the development of novel therapies. Mitochondrial injury in AKI leads to mitochondrial fragmentation and release of damage-associated molecular patterns, which are known to active innate immune pathways and systemic inflammation. This review presents current evidence suggesting that extracellular mitochondrial damage-associated molecular patterns are mediators of remote organ failures during AKI that have the potential to be modifiable.
Collapse
Affiliation(s)
- Mark Hepokoski
- 1Veterans Affairs San Diego Healthcare System, San Diego, California,2Division of Pulmonary and Critical Care Medicine, University of California, San Diego, California
| | - Prabhleen Singh
- 1Veterans Affairs San Diego Healthcare System, San Diego, California,3Division of Nephrology and Hypertension, University of California, San Diego, California
| |
Collapse
|
15
|
Wei X, Wang Y, Weng J, Lao Y, Deng R, Lu J, Yang S, Liu X. Combination of Perindopril Erbumine and Huangqi-Danshen Decoction Protects Against Chronic Kidney Disease via Sirtuin3/Mitochondrial Dynamics Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5812105. [PMID: 35677375 PMCID: PMC9170396 DOI: 10.1155/2022/5812105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/11/2022] [Accepted: 04/27/2022] [Indexed: 11/18/2022]
Abstract
Background Chronic kidney disease (CKD) is a major public health problem worldwide. Treatment with renin-angiotensin system inhibitors can achieve only partial efficacy on renal function decline and renal fibrosis in CKD patients. Huangqi-Danshen decoction (HDD) is a basic Chinese herbal pair which is commonly used to treat CKD with good efficacy. Objectives The current study aimed to investigate the effect of perindopril erbumine (PE), an angiotensin-converting enzyme inhibitor, combined with HDD on adenine-induced CKD rat model and explore the possible mechanism from Sirtuin3/mitochondrial dynamics pathway. Method CKD rat model was established by feeding of 0.75% w/w adenine containing diet for 3 weeks. At the same time, the treatment groups were given PE (0.42 mg/kg/d) or HDD (4.7 g/kg/d) or PE combined with HDD by gavage for 4 weeks. Renal function was evaluated by the levels of serum creatinine (Scr) and blood urea nitrogen (BUN). The renal pathological injury was observed by periodic acid-Schiff (PAS) and Masson's trichrome staining. Proteins expression was determined by Western blot analysis. Mitochondrial morphology was observed by transmission electron microscopy. Results PE in combination with HDD significantly improved renal function, reduced tubular injury and interstitial fibrosis in adenine-induced CKD rats. Moreover, PE + HDD treatment mainly activated the Sirtuin3 expression level. In addition, PE + HDD exhibited bidirectional regulation on mitochondrial dynamics by suppressing mitochondrial fission protein dynaminrelated protein 1 expression and elevating mitochondrial fusion protein optic atrophy 1 expression, resulted in restraint of mitochondrial fragmentation. Conclusion The combination of PE and HDD attenuated adenine-induced CKD in rats, which was possibly associated with Sirtuin3/mitochondrial dynamics pathway.
Collapse
Affiliation(s)
- Xian Wei
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518000, China
| | - Yuzhi Wang
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| | - Jiali Weng
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| | - Yunlan Lao
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| | - Ruyu Deng
- Shenzhen Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Shenzhen 518000, Guangdong, China
| | - Jiandong Lu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518000, China
| | - Shudong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518000, China
| | - Xinhui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518000, China
| |
Collapse
|
16
|
Regulation of Mitochondrial Homeostasis and Nrf2 in Kidney Disease: Timing Is Critical. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9275056. [PMID: 35528519 PMCID: PMC9072027 DOI: 10.1155/2022/9275056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/18/2022] [Indexed: 02/07/2023]
Abstract
Abnormal regulation of mitochondrial homeostasis plays a critical role in the progression of renal disease. Recent studies have shown that activation of nuclear factor erythroid 2-related factor 2 (Nrf2) has time-dependent protective effects, which can be explained by the differing regulation of mitochondrial homeostasis during the various stages of kidney disease. In this review, we summarize the mechanisms whereby mitochondrial homeostasis is regulated and the nature of the dysregulation of mitochondrial homeostasis in renal disease. In addition, we summarize the dual roles of Nrf2 in kidney disease by discussing the studies that have shown the importance of the timing of its activation in the regulation of mitochondrial homeostasis. This should provide a theoretical basis for therapeutic strategies aimed at activating Nrf2 in kidney disease.
Collapse
|
17
|
Bhatia D, Capili A, Nakahira K, Muthukumar T, Torres LK, Choi AMK, Choi ME. Conditional deletion of myeloid-specific mitofusin 2 but not mitofusin 1 promotes kidney fibrosis. Kidney Int 2022; 101:963-986. [PMID: 35227692 PMCID: PMC9038692 DOI: 10.1016/j.kint.2022.01.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/22/2021] [Accepted: 01/11/2022] [Indexed: 11/30/2022]
Abstract
Macrophages exert critical functions during kidney injury, inflammation, and tissue repair or fibrosis. Mitochondrial structural and functional aberrations due to an imbalance in mitochondrial fusion/fission processes are implicated in the pathogenesis of chronic kidney disease. Therefore, we investigated macrophage-specific functions of mitochondrial fusion proteins, mitofusin (MFN)1 and MFN2, in modulating macrophage mitochondrial dynamics, biogenesis, oxidative stress, polarization, and fibrotic response. MFN1 and MFN2 were found to be suppressed in mice after adenine diet-induced chronic kidney disease, in transforming growth factor-beta 1-treated bone marrow-derived macrophages, and in THP-1-derived human macrophages (a human leukemic cell line). However, abrogating Mfn2 but not Mfn1 in myeloid-lineage cells resulted in greater macrophage recruitment into the kidney during fibrosis and the macrophage-derived fibrotic response associated with collagen deposition culminating in worsening kidney function. Myeloid-specific Mfn1 /Mfn2 double knockout mice also showed increased adenine-induced fibrosis. Mfn2-deficient bone marrow-derived macrophages displayed enhanced polarization towards the profibrotic/M2 phenotype and impaired mitochondrial biogenesis. Macrophages in the kidney of Mfn2-deficient and double knockout but not Mfn1-deficient mice exhibited greater mitochondrial mass, size, oxidative stress and lower mitophagy under fibrotic conditions than the macrophages in the kidney of wild-type mice. Thus, downregulation of MFN2 but not MFN1 lead to macrophage polarization towards a profibrotic phenotype to promote kidney fibrosis through a mechanism involving suppression of macrophage mitophagy and dysfunctional mitochondrial dynamics.
Collapse
Affiliation(s)
- Divya Bhatia
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Allyson Capili
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Kiichi Nakahira
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA; Department of Pharmacology, Nara Medical University, Nara, Japan
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA; NewYork-Presbyterian Hospital, New York, New York, USA
| | - Lisa K Torres
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA; NewYork-Presbyterian Hospital, New York, New York, USA
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA; NewYork-Presbyterian Hospital, New York, New York, USA.
| |
Collapse
|
18
|
Li Z, Lu S, Li X. The role of metabolic reprogramming in tubular epithelial cells during the progression of acute kidney injury. Cell Mol Life Sci 2021; 78:5731-5741. [PMID: 34185125 PMCID: PMC11073237 DOI: 10.1007/s00018-021-03892-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/01/2021] [Accepted: 06/25/2021] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is one of the most common clinical syndromes. AKI is associated with significant morbidity and subsequent chronic kidney disease (CKD) development. Thus, it is urgent to develop a strategy to hinder AKI progression. Renal tubules are responsible for the reabsorption and secretion of various solutes and the damage to this part of the nephron is a key mediator of AKI. As we know, many common renal insults primarily target the highly metabolically active proximal tubular cells (PTCs). PTCs are the most energy-demanding cells in the kidney. The ATP that they use is mostly produced in their mitochondria by fatty acid β-oxidation (FAO). But, when PTCs face various biological stresses, FAO will shut down for a time that outlives injury. Recent studies have suggested that surviving PTCs can adapt to FAO disruption by increasing glycolysis when facing metabolic constraints, although PTCs do not perform glycolysis in a normal physiological state. Enhanced glycolysis in a short period compensates for impaired energy production and exerts partial renal-protective effects, but its long-term effect on renal function and AKI progression is not promising. Deranged FAO and enhanced glycolysis may contribute to the AKI to CKD transition through different molecular biological mechanisms. In this review, we concentrate on the recent pathological findings of AKI with regards to the metabolic reprogramming in PTCs, confirming that targeting metabolic reprogramming represents a potentially effective therapeutic strategy for the progression of AKI.
Collapse
Affiliation(s)
- Zhenzhen Li
- Medicial Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Shan Lu
- Emergency Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaobing Li
- College of Basic Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
| |
Collapse
|
19
|
Zheng D, Zhou H, Wang H, Zhu Y, Wu Y, Li Q, Li T, Liu L. Mesenchymal stem cell-derived microvesicles improve intestinal barrier function by restoring mitochondrial dynamic balance in sepsis rats. Stem Cell Res Ther 2021; 12:299. [PMID: 34039427 PMCID: PMC8152336 DOI: 10.1186/s13287-021-02363-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sepsis is a major cause of death in ICU, and intestinal barrier dysfunction is its important complication, while the treatment is limited. Recently, mesenchymal stem cell-derived microvesicles (MMVs) attract much attention as a strategy of cell-free treatment; whether MMVs are therapeutic in sepsis induced-intestinal barrier dysfunction is obscure. METHODS In this study, cecal ligation and puncture-induced sepsis rats and lipopolysaccharide-stimulated intestinal epithelial cells to investigate the effect of MMVs on intestinal barrier dysfunction. MMVs were harvested from mesenchymal stem cells and were injected into sepsis rats, and the intestinal barrier function was measured. Afterward, MMVs were incubated with intestinal epithelial cells, and the effect of MMVs on mitochondrial dynamic balance was measured. Then the expression of mfn1, mfn2, OPA1, and PGC-1α in MMVs were measured by western blot. By upregulation and downregulation of mfn2 and PGC-1α, the role of MMVs in mitochondrial dynamic balance was investigated. Finally, the role of MMV-carried mitochondria in mitochondrial dynamic balance was investigated. RESULTS MMVs restored the intestinal barrier function by improving mitochondrial dynamic balance and metabolism of mitochondria. Further study revealed that MMVs delivered mfn2 and PGC-1α to intestinal epithelial cells, and promoted mitochondrial fusion and biogenesis, thereby improving mitochondrial dynamic balance. Furthermore, MMVs delivered functional mitochondria to intestinal epithelial cells and enhanced energy metabolism directly. CONCLUSION MMVs can deliver mfn2, PGC-1α, and functional mitochondria to intestinal epithelial cells, synergistically improve mitochondrial dynamic balance of target cells after sepsis, and restore the mitochondrial function and intestinal barrier function. The study illustrated that MMVs might be a promising strategy for the treatment of sepsis.
Collapse
Affiliation(s)
- Danyang Zheng
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Daping, Chongqing, 400042, People's Republic of China
| | - Henan Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Daping, Chongqing, 400042, People's Republic of China
| | - Hongchen Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Daping, Chongqing, 400042, People's Republic of China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Daping, Chongqing, 400042, People's Republic of China
| | - Yue Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Daping, Chongqing, 400042, People's Republic of China
| | - Qinghui Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Daping, Chongqing, 400042, People's Republic of China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Daping, Chongqing, 400042, People's Republic of China.
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Daping, Chongqing, 400042, People's Republic of China.
| |
Collapse
|
20
|
Tang C, Cai J, Yin XM, Weinberg JM, Venkatachalam MA, Dong Z. Mitochondrial quality control in kidney injury and repair. Nat Rev Nephrol 2021; 17:299-318. [PMID: 33235391 PMCID: PMC8958893 DOI: 10.1038/s41581-020-00369-0] [Citation(s) in RCA: 292] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 01/30/2023]
Abstract
Mitochondria are essential for the activity, function and viability of eukaryotic cells and mitochondrial dysfunction is involved in the pathogenesis of acute kidney injury (AKI) and chronic kidney disease, as well as in abnormal kidney repair after AKI. Multiple quality control mechanisms, including antioxidant defence, protein quality control, mitochondrial DNA repair, mitochondrial dynamics, mitophagy and mitochondrial biogenesis, have evolved to preserve mitochondrial homeostasis under physiological and pathological conditions. Loss of these mechanisms may induce mitochondrial damage and dysfunction, leading to cell death, tissue injury and, potentially, organ failure. Accumulating evidence suggests a role of disturbances in mitochondrial quality control in the pathogenesis of AKI, incomplete or maladaptive kidney repair and chronic kidney disease. Moreover, specific interventions that target mitochondrial quality control mechanisms to preserve and restore mitochondrial function have emerged as promising therapeutic strategies to prevent and treat kidney injury and accelerate kidney repair. However, clinical translation of these findings is challenging owing to potential adverse effects, unclear mechanisms of action and a lack of knowledge of the specific roles and regulation of mitochondrial quality control mechanisms in kidney resident and circulating cell types during injury and repair of the kidney.
Collapse
Affiliation(s)
- Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Juan Cai
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel M. Weinberg
- Department of Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Manjeri A. Venkatachalam
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital at Central South University, Changsha, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA.,
| |
Collapse
|
21
|
Giraud S, Kerforne T, Zely J, Ameteau V, Couturier P, Tauc M, Hauet T. The inhibition of eIF5A hypusination by GC7, a preconditioning protocol to prevent brain death-induced renal injuries in a preclinical porcine kidney transplantation model. Am J Transplant 2020; 20:3326-3340. [PMID: 32400964 DOI: 10.1111/ajt.15994] [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: 02/13/2020] [Revised: 04/07/2020] [Accepted: 04/29/2020] [Indexed: 01/25/2023]
Abstract
The eIF5A hypusination inhibitor GC7 (N1-guanyl-1,7-diaminoheptane) was shown to protect from ischemic injuries. We hypothesized that GC7 could be useful for preconditioning kidneys from donors before transplantation. Using a preclinical porcine brain death (BD) donation model, we carried out in vivo evaluation of GC7 pretreatment (3 mg/kg iv, 5 minutes after BD) at the beginning of the 4h-donor management, after which kidneys were collected and cold-stored (18h in University of Wisconsin solution) and 1 was allotransplanted. Groups were defined as following (n = 6 per group): healthy (CTL), untreated BD (Vehicle), and GC7-treated BD (Vehicle + GC7). At the end of 4h-management, GC7 treatment decreased BD-induced markers, as radical oxygen species markers. In addition, GC7 increased expression of mitochondrial protective peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC1α) and antioxidant proteins (superoxyde-dismutase-2, heme oxygenase-1, nuclear factor [erythroid-derived 2]-like 2 [NRF2], and sirtuins). At the end of cold storage, GC7 treatment induced an increase of NRF2 and PGC1α mRNA and a better mitochondrial integrity/homeostasis with a decrease of dynamin- related protein-1 activation and increase of mitofusin-2. Moreover, GC7 treatment significantly improved kidney outcome during 90 days follow-up after transplantation (fewer creatininemia and fibrosis). Overall, GC7 treatment was shown to be protective for kidneys against BD-induced injuries during donor management and subsequently appeared to preserve antioxidant defenses and mitochondria homeostasis; these protective effects being accompanied by a better transplantation outcome.
Collapse
Affiliation(s)
- Sebastien Giraud
- INSERM UMR-1082 IRTOMIT, Poitiers, France.,Service de Biochimie, CHU de Poitiers, Poitiers, France
| | - Thomas Kerforne
- INSERM UMR-1082 IRTOMIT, Poitiers, France.,Faculté de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France.,Service d'Anesthésie-Réanimation, CHU de Poitiers, Poitiers, France
| | - Jeremy Zely
- INSERM UMR-1082 IRTOMIT, Poitiers, France.,Faculté de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France.,Service d'Anesthésie-Réanimation, CHU de Poitiers, Poitiers, France
| | - Virginie Ameteau
- INSERM UMR-1082 IRTOMIT, Poitiers, France.,Faculté de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France
| | - Pierre Couturier
- INSERM UMR-1082 IRTOMIT, Poitiers, France.,Service de Biochimie, CHU de Poitiers, Poitiers, France.,MOPICT 'plate-forme MOdélisation Préclinique - Innovations Chirurgicale et Technologique', Domaine Expérimental du Magneraud, Surgères, France
| | - Michel Tauc
- Université Cote d'Azur, LP2M, CNRS-7370, Nice, France
| | - Thierry Hauet
- INSERM UMR-1082 IRTOMIT, Poitiers, France.,Service de Biochimie, CHU de Poitiers, Poitiers, France.,Faculté de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France.,MOPICT 'plate-forme MOdélisation Préclinique - Innovations Chirurgicale et Technologique', Domaine Expérimental du Magneraud, Surgères, France.,FHU SUPORT 'SUrvival oPtimization in ORgan Transplantation', Poitiers, France
| |
Collapse
|
22
|
Jiang M, Bai M, Lei J, Xie Y, Xu S, Jia Z, Zhang A. Mitochondrial dysfunction and the AKI-to-CKD transition. Am J Physiol Renal Physiol 2020; 319:F1105-F1116. [PMID: 33073587 DOI: 10.1152/ajprenal.00285.2020] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Acute kidney injury (AKI) has been widely recognized as an important risk factor for the occurrence and development of chronic kidney disease (CKD). Even milder AKI has adverse consequences and could progress to renal fibrosis, which is the ultimate common pathway for various terminal kidney diseases. Thus, it is urgent to develop a strategy to hinder the transition from AKI to CKD. Some mechanisms of the AKI-to-CKD transition have been revealed, such as nephron loss, cell cycle arrest, persistent inflammation, endothelial injury with vascular rarefaction, and epigenetic changes. Previous studies have elucidated the pivotal role of mitochondria in acute injuries and demonstrated that the fitness of this organelle is a major determinant in both the pathogenesis and recovery of organ function. Recent research has suggested that damage to mitochondrial function in early AKI is a crucial factor leading to tubular injury and persistent renal insufficiency. Dysregulation of mitochondrial homeostasis, alterations in bioenergetics, and organelle stress cross talk contribute to the AKI-to-CKD transition. In this review, we focus on the pathophysiology of mitochondria in renal recovery after AKI and progression to CKD, confirming that targeting mitochondria represents a potentially effective therapeutic strategy for the progression of AKI to CKD.
Collapse
Affiliation(s)
- Mingzhu Jiang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Mi Bai
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Lei
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yifan Xie
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Shuang Xu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
23
|
Rahbar Saadat Y, Hosseiniyan Khatibi SM, Ardalan M, Barzegari A, Zununi Vahed S. Molecular pathophysiology of acute kidney injury: The role of sirtuins and their interactions with other macromolecular players. J Cell Physiol 2020; 236:3257-3274. [PMID: 32989772 DOI: 10.1002/jcp.30084] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
Acute kidney injury (AKI), a rapid drop in kidney function, displays high mortality and morbidity, and its repeated or severe status can shift into chronic kidney disease or even end-stage renal disease. How and which events cause AKI still is controversial. In addition, no specific therapies have emerged that can attenuate AKI or expedite recovery. Some central mechanisms including tubular epithelial cells injury, endothelial injury, renal cell apoptosis, and necrosis signaling cascades, and inflammation have been reported in the pathophysiology of AKI. However, the timing of the activation of each pathway, their interactions, and the hierarchy of these pathways remain unknown. The main molecular mechanisms that might be complicated in this process are the mitochondrial impairment and alteration/shifting of cellular metabolites (e.g., acetyl-CoA and NAD+ /NADH) acting as cofactors to alter the activities of many enzymes, for instance, sirtuins. Moreover, alteration of mitochondrial structure over the fusion and fission mechanisms can regulate cellular signaling pathways by modifying the rate of reactive oxygen species generation and metabolic activities. The aim of this review is to better understand the underlying pathophysiological and molecular mechanisms of AKI. In addition, we predicted the main other molecular players in interaction with sirtuins as energy/stresses monitoring proteins for the development of future approaches in the treatment or prevention of ischemic AKI.
Collapse
Affiliation(s)
- Yalda Rahbar Saadat
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.,INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Université Sorbonne Paris Nord, Villetaneuse, France
| | | |
Collapse
|
24
|
Agil A, Chayah M, Visiedo L, Navarro-Alarcon M, Rodríguez Ferrer JM, Tassi M, Reiter RJ, Fernández-Vázquez G. Melatonin Improves Mitochondrial Dynamics and Function in the Kidney of Zücker Diabetic Fatty Rats. J Clin Med 2020; 9:jcm9092916. [PMID: 32927647 PMCID: PMC7564180 DOI: 10.3390/jcm9092916] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity and associated diabetes (diabesity) impair kidney mitochondrial dynamics by augmenting fission and diminishing fusion, which results in mitochondrial and renal dysfunction. Based on available evidence, the antioxidant activities of melatonin may improve impaired renal mitochondrial function in obese diabetic animals by restoring the imbalanced dynamics through inhibiting fission and promoting fusion. Male Zücker diabetic fatty (ZDF) rats and lean littermates (ZL) were orally treated either with melatonin (10 mg/kg BW/day) (M-ZDF and M-ZL) or vehicle (C-ZDF and C-ZL) for 17 weeks. Kidney function was evaluated by measurement of total urine volume, proteinuria, creatinine clearance, and assessment of kidney mitochondrial dynamics and function. C-ZDF exhibited impaired dynamics and function of kidney mitochondria in comparison to C-ZL. Melatonin improved nephropathy of ZDF rats and modulated their mitochondrial dynamics by reducing expression of Drp1 fission marker and increasing that of fusion markers, Mfn2 and Opa1. Furthermore, melatonin ameliorated mitochondrial dysfunction by increasing respiratory control index and electron transfer chain complex IV activity. In addition, it lowered mitochondrial oxidative status. Our findings show that melatonin supplementation improves nephropathy likely via modulation of the mitochondrial fission/fusion balance and function in ZDF rats.
Collapse
Affiliation(s)
- Ahmad Agil
- Department of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, 18016 Granada, Spain; (M.C.); (L.V.)
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospital of Granada, 18016 Granada, Spain
- Correspondence: ; Tel.: +34-625-143-359
| | - Meriem Chayah
- Department of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, 18016 Granada, Spain; (M.C.); (L.V.)
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospital of Granada, 18016 Granada, Spain
| | - Lucia Visiedo
- Department of Pharmacology and Neurosciences Institute, School of Medicine, University of Granada, 18016 Granada, Spain; (M.C.); (L.V.)
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospital of Granada, 18016 Granada, Spain
| | - Miguel Navarro-Alarcon
- Department of Nutrition and Bromatology, School of Pharmacy, University of Granada, 18071 Granada, Spain;
| | | | - Mohamed Tassi
- Service of Microscopy, CIBM, University of Granada, 18016 Granada, Spain;
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science at San Antonio, San Antonio, TX 78229, USA;
| | | |
Collapse
|
25
|
Abstract
Mitochondria fulfill the high metabolic energy demands of the kidney and are regularly exposed to oxidative stress causing mitochondrial damage. The selective removal of damaged and dysfunctional mitochondria through a process known as mitophagy is essential in maintaining cellular homeostasis and physiological function. Mitochondrial quality control by mitophagy is particularly crucial for an organ such as the kidney, which is rich in mitochondria. The role of mitophagy in the pathogenesis of kidney diseases has lately gained significant attention. In this review, we summarize the current understanding of the implications of mitophagy during pathological conditions of the kidney, including acute and chronic kidney diseases.
Collapse
Affiliation(s)
- Divya Bhatia
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
26
|
Wang Z, Salih E, Igwebuike C, Mulhern R, Bonegio RG, Havasi A, Borkan SC. Nucleophosmin Phosphorylation as a Diagnostic and Therapeutic Target for Ischemic AKI. J Am Soc Nephrol 2019; 30:50-62. [PMID: 30573638 DOI: 10.1681/asn.2018040401] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 10/18/2018] [Indexed: 02/04/2023] Open
Abstract
Background Ischemic AKI lacks a urinary marker for early diagnosis and an effective therapy. Differential nucleophosmin (NPM) phosphorylation is a potential early marker of ischemic renal cell injury and a therapeutic target.Methods Differential NPM phosphorylation was assessed by mass spectrometry in NPM harvested from murine and human primary renal epithelial cells, fresh kidney tissue, and urine before and after ischemic injury. The biologic behavior and toxicity of NPM was assessed using phospho-NPM mutant proteins that either mimic stress-induced or normal NPM phosphorylation. Peptides designed to interfere with NPM function were used to explore NPM as a therapeutic target.Results Within hours of stress, virtually identical phosphorylation changes were detected at distinct serine/threonine sites in NPM harvested from primary renal cells, tissue, and urine. A phosphomimic NPM protein that replicated phosphorylation under stress localized to the cytosol, formed monomers that interacted with Bax, a cell death protein, coaccumulated with Bax in isolated mitochondria, and significantly increased cell death after stress; wild-type NPM or a phosphomimic NPM with a normal phosphorylation configuration did not. Three renal targeted peptides designed to interfere with NPM at distinct functional sites significantly protected against cell death, and a single dose of one peptide administered several hours after ischemia that would be lethal in untreated mice significantly reduced AKI severity and improved survival.Conclusions These findings establish phosphorylated NPM as a potential early marker of ischemic AKI that links early diagnosis with effective therapeutic interventions.
Collapse
Affiliation(s)
- Zhiyong Wang
- Renal Section, Boston University Medical Center, Boston, Massachusetts; and
| | - Erdjan Salih
- Department of Periodontology, Goldman School of Dentistry, Boston University, Boston, Massachusetts
| | | | - Ryan Mulhern
- Renal Section, Boston University Medical Center, Boston, Massachusetts; and
| | - Ramon G Bonegio
- Renal Section, Boston University Medical Center, Boston, Massachusetts; and
| | - Andrea Havasi
- Renal Section, Boston University Medical Center, Boston, Massachusetts; and
| | - Steven C Borkan
- Renal Section, Boston University Medical Center, Boston, Massachusetts; and
| |
Collapse
|
27
|
Mitochondrial dynamics and their potential as a therapeutic target. Mitochondrion 2019; 49:269-283. [PMID: 31228566 DOI: 10.1016/j.mito.2019.06.002] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/02/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022]
Abstract
Mitochondrial dynamics shape the mitochondrial network and contribute to mitochondrial function and quality control. Mitochondrial fusion and division are integrated into diverse cellular functions and respond to changes in cell physiology. Imbalanced mitochondrial dynamics are associated with a range of diseases that are broadly characterized by impaired mitochondrial function and increased cell death. In various disease models, modulating mitochondrial fusion and division with either small molecules or genetic approaches has improved function. Although additional mechanistic understanding of mitochondrial fusion and division will be critical to inform further therapeutic approaches, mitochondrial dynamics represent a powerful therapeutic target in a wide range of human diseases.
Collapse
|
28
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Mesenchymal stem cell therapy targeting mitochondrial dysfunction in acute kidney injury. J Transl Med 2019; 17:142. [PMID: 31046805 PMCID: PMC6498508 DOI: 10.1186/s12967-019-1893-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/25/2019] [Indexed: 12/13/2022] Open
Abstract
Mitochondria take part in a network of cellular processes that regulate cell homeostasis. Defects in mitochondrial function are key pathophysiological changes during acute kidney injury (AKI). Mesenchymal stem cells (MSCs) have shown promising regenerative effects in experimental AKI models, but the specific mechanism is still unclear. Some studies have demonstrated that MSCs are able to target mitochondrial dysfunction during AKI. In this review, we summarize these articles, providing an integral and updated view of MSC therapy targeting mitochondrial dysfunction during AKI, which is aimed at promoting the therapeutic effect of MSCs in AKI patients.
Collapse
Affiliation(s)
- Lingfei Zhao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ping Zhang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hua Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China. .,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
29
|
Cameron RB, Gibbs WS, Miller SR, Dupre TV, Megyesi J, Beeson CC, Schnellmann RG. Proximal Tubule β 2-Adrenergic Receptor Mediates Formoterol-Induced Recovery of Mitochondrial and Renal Function after Ischemia-Reperfusion Injury. J Pharmacol Exp Ther 2019; 369:173-180. [PMID: 30709866 PMCID: PMC11046739 DOI: 10.1124/jpet.118.252833] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 01/04/2019] [Indexed: 04/28/2024] Open
Abstract
Acute kidney injury (AKI) is the rapid loss of renal function after an insult, and renal proximal tubule cells (RPTCs) are central to the pathogenesis of AKI. The β 2-adrenergic receptor (β 2AR) agonist formoterol accelerates the recovery of renal function in mice after ischemia-reperfusion injury (IRI) with associated rescue of mitochondrial proteins; however, the cell type responsible for this recovery remains unknown. The role of RPTCs in formoterol-induced recovery of renal function was assessed in a proximal tubule-specific knockout of the β 2AR (γGT-Cre:ADRB2Flox/Flox). These mice and wild-type controls (ADRB2Flox/Flox) were subjected to renal IRI, followed by once-daily dosing of formoterol beginning 24 hours post-IRI and euthanized at 144 hours. Compared with ADRB2Flox/Flox mice, γGT-Cre:ADRB2Flox/Flox mice had decreased renal cortical mRNA expression of the β 2AR. After IRI, formoterol treatment restored renal function in ADRB2Flox/Flox but not γGT-Cre:ADRB2Flox/Flox mice as measured by serum creatinine, histopathology, and expression of kidney injury marker-1 (KIM-1). Formoterol-treated ADRB2Flox/Flox mice exhibited recovery of mitochondrial proteins and DNA copy number, whereas γGT-Cre:ADRB2Flox/Flox mice treated with formoterol did not. Analysis of mitochondrial morphology by transmission electron microscopy demonstrated that formoterol increased mitochondrial number and density in ADRB2Flox/Flox mice but not in γGT-Cre:ADRB2Flox/Flox mice. These data demonstrate that proximal tubule β 2AR regulates renal mitochondrial homeostasis. Formoterol accelerates the recovery of renal function after AKI by activating proximal tubule β 2AR to induce mitochondrial biogenesis and demonstrates the overall requirement of RPTCs in renal recovery.
Collapse
Affiliation(s)
- Robert B Cameron
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (R.B.C., W.S.G., C.C.B.); College of Pharmacy, University of Arizona (R.B.C., W.S.G., S.R.M., T.V.D., R.G.S.), and Southern Arizona VA Healthcare System (R.G.S.), Tuscon, Arizona; and Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas (J.M.)
| | - Whitney S Gibbs
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (R.B.C., W.S.G., C.C.B.); College of Pharmacy, University of Arizona (R.B.C., W.S.G., S.R.M., T.V.D., R.G.S.), and Southern Arizona VA Healthcare System (R.G.S.), Tuscon, Arizona; and Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas (J.M.)
| | - Siennah R Miller
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (R.B.C., W.S.G., C.C.B.); College of Pharmacy, University of Arizona (R.B.C., W.S.G., S.R.M., T.V.D., R.G.S.), and Southern Arizona VA Healthcare System (R.G.S.), Tuscon, Arizona; and Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas (J.M.)
| | - Tess V Dupre
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (R.B.C., W.S.G., C.C.B.); College of Pharmacy, University of Arizona (R.B.C., W.S.G., S.R.M., T.V.D., R.G.S.), and Southern Arizona VA Healthcare System (R.G.S.), Tuscon, Arizona; and Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas (J.M.)
| | - Judit Megyesi
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (R.B.C., W.S.G., C.C.B.); College of Pharmacy, University of Arizona (R.B.C., W.S.G., S.R.M., T.V.D., R.G.S.), and Southern Arizona VA Healthcare System (R.G.S.), Tuscon, Arizona; and Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas (J.M.)
| | - Craig C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (R.B.C., W.S.G., C.C.B.); College of Pharmacy, University of Arizona (R.B.C., W.S.G., S.R.M., T.V.D., R.G.S.), and Southern Arizona VA Healthcare System (R.G.S.), Tuscon, Arizona; and Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas (J.M.)
| | - Rick G Schnellmann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (R.B.C., W.S.G., C.C.B.); College of Pharmacy, University of Arizona (R.B.C., W.S.G., S.R.M., T.V.D., R.G.S.), and Southern Arizona VA Healthcare System (R.G.S.), Tuscon, Arizona; and Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas (J.M.)
| |
Collapse
|
30
|
Aydogan Mathyk B, Temel Yuksel I, Tayyar A, Aslan Cetin B, Tayyar AT, Koroglu N. Maternal serum mitofusin-2 levels in patients with preeclampsia: the possible role of mitochondrial dysfunction in preeclampsia. J Matern Fetal Neonatal Med 2019; 33:1861-1866. [PMID: 30614327 DOI: 10.1080/14767058.2018.1532497] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Purpose: Hypoxia alters mitochondria function and our aim was to measure mitochondrial fusion protein mitofusin-2 (Mfn2) in patients with preeclampsia.Materials and methods: This cross-sectional study was conducted including 82 pregnant women, 27 with normal pregnancy and 55 with preeclampsia (27 with early-onset preeclampsia and 28 with late-onset preeclampsia). Maternal serum levels of Mfn2 were measured by using enzyme-linked immunosorbent assay kits.Results: The mean serum mitofusin-2 levels were higher in women with preeclampsia than in the control group (68.02 ± 8.7 pg/mL vs. 99.72 ± 37.27 pg/mL, p < .0001). The mean serum mitofusin-2 level was found to be the highest in the early-onset preeclampsia (EOPE) group (EOPE: 101.6 ± 38.5 pg/mL). Maternal serum mitofusin-2 levels correlated with both systolic and diastolic blood pressures as well as uterine artery pulsatility index. The optimal cutoff value of Mfn2 for determining preeclampsia was 75.3 pg/mL.Conclusion: Mfn2 has regulatory roles in stress response. Maternal serum Mfn2 is higher in patients with preeclampsia suggesting that Mfn2 increases in the maternal system as a stress response against hypoxia and endothelial dysfunction.What do the results of this study add? Hypoxia causes mitochondrial dysfunction that has been linked to the etiology of many diseases including preeclampsia. Mitofusin-2 is a mitochondrial fusion protein, and the levels can be altered in preeclampsia. For the first time, we showed that maternal levels of mitofusin-2 are higher in patients with preeclampsia. Further, we reported the correlation of mitofusin-2 with blood pressures and uterine artery pulsatility index. These findings will open up other avenues for researchers to investigate other mitochondrial molecules while under stress.
Collapse
Affiliation(s)
- Begum Aydogan Mathyk
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of North Carolina, Chapel Hill, NC, USA
| | - Ilkbal Temel Yuksel
- Department of Obstetrics and Gynecology and Kanuni Sultan Suleyman Research and Training Hospital, Istanbul, Turkey
| | - Ahmet Tayyar
- Department of Obstetrics and Gynecology and Kanuni Sultan Suleyman Research and Training Hospital, Istanbul, Turkey
| | - Berna Aslan Cetin
- Department of Obstetrics and Gynecology and Kanuni Sultan Suleyman Research and Training Hospital, Istanbul, Turkey
| | - Ahter Tanay Tayyar
- Department of Obstetrics and Gynecology, Health Sciences University, Zeynep Kamil Research and Training Hospital, Istanbul, Turkey
| | - Nadiye Koroglu
- Department of Obstetrics and Gynecology and Kanuni Sultan Suleyman Research and Training Hospital, Istanbul, Turkey
| |
Collapse
|
31
|
Huangqi-Danshen Decoction Ameliorates Adenine-Induced Chronic Kidney Disease by Modulating Mitochondrial Dynamics. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9574045. [PMID: 30713579 PMCID: PMC6332985 DOI: 10.1155/2019/9574045] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/05/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is a leading public health problem with high morbidity and mortality. However, the therapies remain limited. Traditional Chinese medicine (TCM) has been used for treating kidney disease for thousands of years and is an effective alternative treatment for CKD patients in China and other Asian countries. In the present study, we aimed to investigate the effect and mechanism of Huangqi-Danshen decoction (HDD), a TCM herbal decoction, on treating CKD. CKD rat model was induced by adding 0.75% adenine to the diet for 4 weeks. HDD extract was administrated orally to CKD rats at the dose of 4.7 g/kg/d for consecutive 4 weeks in adenine-induced CKD rats. Kidney function was evaluated by the levels of serum creatinine (Scr) and blood urea nitrogen (BUN). The pathological changes of kidney tissues were observed by periodic acid-Schiff (PAS) and Masson's trichrome staining. The proteins expression of renal fibrosis and mitochondrial dynamics were determined and quantified by Western blot analysis. CKD rats showed obvious decline in renal function as evidenced by increased levels of Scr and BUN, which were blunted by HDD treatment. HDD could also improve tubular atrophy and interstitial fibrosis of CKD rats. Moreover, HDD downregulated fibronectin, type IV collagen, and α-smooth muscle actin expression in CKD rats. Furthermore, mitochondrial dynamics was disturbed in CKD rats, which manifested as increased mitochondrial fission and decreased mitochondrial fusion. HDD treatment restored mitochondrial dynamics in CKD rats by repressing dynamin-related protein 1 and Mid 49/51 expression, promoting mitofusin 2 expression, and suppressing optic atrophy 1 proteolysis. In conclusion, HDD could significantly retard CKD progression through modulating mitochondrial dynamics.
Collapse
|
32
|
Tsushida K, Tanabe K, Masuda K, Tanimura S, Miyake H, Arata Y, Sugiyama H, Wada J. Estrogen-related receptor α is essential for maintaining mitochondrial integrity in cisplatin-induced acute kidney injury. Biochem Biophys Res Commun 2018; 498:918-924. [PMID: 29545177 DOI: 10.1016/j.bbrc.2018.03.080] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 03/10/2018] [Indexed: 11/24/2022]
Abstract
Acute kidney injury (AKI) has been associated with not only higher in-hospital mortality but also the subsequent development of chronic kidney disease (CKD). Recent evidence has suggested the involvement of mitochondrial dysfunction and impaired dynamics in the pathogenesis of AKI. Estrogen-related receptor α (ERRα) is an orphan nuclear receptor that acts as a transcription factor to regulate the transcription of genes required for mitochondrial biogenesis and oxidative phosphorylation. In the present study, we examined the effects of ERRα deficiency on the progression of AKI induced by cisplatin. Male C57BL/6 J wild-type and ERRα-/- mice received a single intraperitoneal injection of 20 mg/kg cisplatin. Seventy-two hours after the injection, kidney function and morphology were evaluated. ERRα expression was observed in renal tubules, and cisplatin inhibited its translocation into nuclei. ERRα deficiency exacerbated cisplatin-induced renal dysfunction and tubular injury, as well as oxidative stress and apoptosis. ERRα-/- mice kidneys revealed lower mitochondrial DNA content and swollen mitochondria with reduced cristae. In addition, these mice had lower expression of the mitochondrial fusion protein mitofusin-2. The cisplatin-induced decrease in mitochondrial DNA and altered mitochondrial structure were more severe in ERRα-/- mice. In cultured mouse proximal tubular epithelial cells, the ERRα inverse agonist XCT-790 significantly inhibited mitofusin-2 expression and induced mitochondrial fragmentation. Taken together, our findings suggest the involvement of ERRα in the progression of cisplatin-induced AKI probably through impaired mitochondrial dynamics.
Collapse
Affiliation(s)
- Keigo Tsushida
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Tanabe
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Kana Masuda
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Tanimura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromasa Miyake
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuka Arata
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hitoshi Sugiyama
- Department of Human Resource Development of Dialysis Therapy for Kidney Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
33
|
Shin JK, Lee SM. Genipin protects the liver from ischemia/reperfusion injury by modulating mitochondrial quality control. Toxicol Appl Pharmacol 2017; 328:25-33. [DOI: 10.1016/j.taap.2017.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 12/26/2022]
|
34
|
Liu N, Jiang Z, Liu Y, Nie Y, Chen J, Ouyang B, Guan X, Chen M. Human trypsin inhibitor reduces the apoptosis of lipopolysaccharide‑induced human kidney‑2 cells by promoting mitochondrial fusion. Mol Med Rep 2017; 16:2899-2906. [PMID: 28713932 DOI: 10.3892/mmr.2017.6927] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 05/02/2017] [Indexed: 11/05/2022] Open
Abstract
Imbalance in mitochondrial fusion/fission is one of the mechanisms leading to sepsis‑induced mitochondrial dysfunction and cell apoptosis. The present study examined the effects of human trypsin inhibitor (UTI), a well‑known antioxidant and anti‑inflammatory substance, on mitochondrial dynamics and cell apoptosis in lipopolysaccharide (LPS)‑induced human kidney‑2 (HK‑2) cells. The HK‑2 cells were incubated for 24 h either with LPS (800 ng/ml) or LPS (800 ng/ml) mixed with UTI (250 U/ml). Cell viability was assessed using a3‑(4,5‑dimethyl‑2‑thiazolyl)‑2, 5‑diphenyl‑2‑H‑tetrazolium bromide assay. Oxidative activities (estimated by maleic dialdehyde and superoxide dismutase), levels of inflammatory cytokines interleukin (IL)‑6 and tumor necrosis factor (TNF)‑α, and levels of ATP were measured using an enzyme‑linked immunosorbent assay. The expression levels of the mitochondrial fission protein, death‑associated protein kinase 2 (DAPK‑2), mitofusin (Mfn)1 and Mfn2 mitochondrial fusion proteins, and apoptotic and anti‑apoptotic biomarkers, including cytochrome c, caspase‑3, caspase‑9, B‑cell lymphoma (Bcl)‑2, Bcl‑extra large and poly ADP‑ribose polymerase (PARP), were assessed using western blot analyses. The changes in mitochondrial membrane potential were analyzed following JC‑1 staining. Annexin V/propidium iodide assays were used to evaluate cell apoptosis. The results showed that the balance of mitochondrial dynamics was towards mitochondrial fusion in the UTI group, as a reduced expression of DAPK2, and increased expression levels of Mfn1 and Mfn2 were detected (P<0.05, vs. LPS group). In addition, adecline in the levels of the inflammatory cytokines, TNF‑α and IL‑6, and the oxidative activities, reflected by an increase in SOD and a decrease in MDA (P<0.05, vs. LPS group) were observed. Cell apoptosis was inhibited following co‑treatment with UTI (P<0.05, vs. LPS group). It was concluded that UTI may protect mitochondrial functions by promoting mitochondrial fusion and limiting mitochondrial fission, thus reducing the apoptosis of LPS‑induced HK‑2 cells.
Collapse
Affiliation(s)
- Ning Liu
- Department of Surgical Intensive Care Unit of The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhiyi Jiang
- Department of Surgical Intensive Care Unit of The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yongjun Liu
- Department of Surgical Intensive Care Unit of The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yao Nie
- Department of Surgical Intensive Care Unit of The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Juan Chen
- Department of Surgical Intensive Care Unit of The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Bin Ouyang
- Department of Surgical Intensive Care Unit of The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiangdong Guan
- Department of Surgical Intensive Care Unit of The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Minying Chen
- Department of Surgical Intensive Care Unit of The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
35
|
Liu X, Yang J, Lu C, Jiang S, Nie X, Han J, Yin L, Jiang J. Downregulation of Mfn2 participates in manganese-induced neuronal apoptosis in rat striatum and PC12 cells. Neurochem Int 2017; 108:40-51. [PMID: 28232070 DOI: 10.1016/j.neuint.2017.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 12/24/2022]
Abstract
Manganese (Mn) is a widely distributed trace element that is essential for normal brain function and development. However, chronic exposure to excessive Mn has been known to lead to neuronal loss and manganism, a disease with debilitating motor and cognitive deficits, whose clinical syndrome resembling idiopathic Parkinson's disease (IPD). However, the precise molecular mechanism underlying Mn neurotoxicity remains largely unclear. Accumulating evidence indicates that abnormal mitochondrial functionality is an early and causal event in Mn-induced neurodegeneration and apoptosis. Here, we investigated whether Mitofusin 2 (Mfn2), a highly conserved dynamin-related protein (DRP), played a role in the regulation of Mn-induced neuronal apoptosis. We revealed that Mfn2 was significantly dysregulated in rat striatum and PC12 neuronal-like cells following Mn exposure. Western blot analysis revealed that the expression of Mfn2 was remarkably decreased following different concentrations of Mn exposure. Immunohistochemistry analysis confirmed a remarkable downregulation of Mfn2 in rat striatum after Mn exposure. Immunofluorescent staining showed that Mfn2 was expressed predominantly in neurons, and neuronal loss of Mfn2 was associated with the expression of active caspase-3 following Mn exposure. Importantly, overexpression of Mfn2 apparently attenuated Mn-induced neuronal apoptosis. Notably, treatment with caspase-3 inhibitor Ac-DEVD-CH could not rescue Mn-induced downregulation of Mfn2, suggesting that Mn-induced mfn2 occurs prior to neuronal apoptosis. Taken together, these results indicated that down-regulated expression of Mfn2 might contribute to the pathological processes underlying Mn neurotoxicity.
Collapse
Affiliation(s)
- Xinhang Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Jianbin Yang
- Department of Public Health, The Second People's Hospital of Nantong, Nantong, Jiangsu Province, People's Republic of China
| | - Chunhua Lu
- Department of Occupational Health and Occupational Diseases, Nantong Center for Disease Control and Prevention, Nantong, Jiangsu Province, People's Republic of China
| | - Shengyang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Xiaoke Nie
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Jingling Han
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Lifeng Yin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China.
| |
Collapse
|
36
|
Pedraza-Chaverri J, Sánchez-Lozada LG, Osorio-Alonso H, Tapia E, Scholze A. New Pathogenic Concepts and Therapeutic Approaches to Oxidative Stress in Chronic Kidney Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6043601. [PMID: 27429711 PMCID: PMC4939360 DOI: 10.1155/2016/6043601] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/16/2016] [Accepted: 05/25/2016] [Indexed: 12/24/2022]
Abstract
In chronic kidney disease inflammatory processes and stimulation of immune cells result in overproduction of free radicals. In combination with a reduced antioxidant capacity this causes oxidative stress. This review focuses on current pathogenic concepts of oxidative stress for the decline of kidney function and development of cardiovascular complications. We discuss the impact of mitochondrial alterations and dysfunction, a pathogenic role for hyperuricemia, and disturbances of vitamin D metabolism and signal transduction. Recent antioxidant therapy options including the use of vitamin D and pharmacologic therapies for hyperuricemia are discussed. Finally, we review some new therapy options in diabetic nephropathy including antidiabetic agents (noninsulin dependent), plant antioxidants, and food components as alternative antioxidant therapies.
Collapse
Affiliation(s)
| | - Laura G. Sánchez-Lozada
- Laboratory of Renal Physiopathology, INC Ignacio Chávez, 14080 Mexico City, DF, Mexico
- Department of Nephrology, INC Ignacio Chávez, 14080 Mexico City, DF, Mexico
| | - Horacio Osorio-Alonso
- Laboratory of Renal Physiopathology, INC Ignacio Chávez, 14080 Mexico City, DF, Mexico
- Department of Nephrology, INC Ignacio Chávez, 14080 Mexico City, DF, Mexico
| | - Edilia Tapia
- Laboratory of Renal Physiopathology, INC Ignacio Chávez, 14080 Mexico City, DF, Mexico
- Department of Nephrology, INC Ignacio Chávez, 14080 Mexico City, DF, Mexico
| | - Alexandra Scholze
- Department of Nephrology, Odense University Hospital, 5000 Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
37
|
Agarwal A, Dong Z, Harris R, Murray P, Parikh SM, Rosner MH, Kellum JA, Ronco C. Cellular and Molecular Mechanisms of AKI. J Am Soc Nephrol 2016; 27:1288-99. [PMID: 26860342 DOI: 10.1681/asn.2015070740] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In this article, we review the current evidence for the cellular and molecular mechanisms of AKI, focusing on epithelial cell pathobiology and related cell-cell interactions, using ischemic AKI as a model. Highlighted are the clinical relevance of cellular and molecular targets that have been investigated in experimental models of ischemic AKI and how such models might be improved to optimize translation into successful clinical trials. In particular, development of more context-specific animal models with greater relevance to human AKI is urgently needed. Comorbidities that could alter patient susceptibility to AKI, such as underlying diabetes, aging, obesity, cancer, and CKD, should also be considered in developing these models. Finally, harmonization between academia and industry for more clinically relevant preclinical testing of potential therapeutic targets and better translational clinical trial design is also needed to achieve the goal of developing effective interventions for AKI.
Collapse
Affiliation(s)
- Anupam Agarwal
- Division of Nephrology, and Nephrology Research and Training Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama;
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, Georgia
| | - Raymond Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Patrick Murray
- Department of Medicine, University College of Dublin, Dublin, Ireland
| | - Samir M Parikh
- Division of Nephrology and Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Mitchell H Rosner
- Department of Medicine, Nephrology Division, and the Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - John A Kellum
- Center for Critical Care Nephrology, Clinical Research, Investigation and Systems Modeling of Acute Illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Claudio Ronco
- Department of Nephrology, Dialysis, and Transplantation, San Bortolo Hospital, and the International Renal Research Institute, 36100 Vicenza, Italy
| | | |
Collapse
|
38
|
Downregulation of Mitofusin 2 in Placenta Is Related to Preeclampsia. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6323086. [PMID: 26942197 PMCID: PMC4752976 DOI: 10.1155/2016/6323086] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 12/01/2015] [Accepted: 12/02/2015] [Indexed: 12/21/2022]
Abstract
Background. Mitofusin 2 (Mfn2) is a novel mitochondrial protein that is implicated in cellular proliferation and metabolism; however, the role of Mfn2 in preeclampsia (PE) remains unknown. This study aimed to explore the relationship between Mfn2 and PE. Method. Preeclamptic and normal pregnancies were enrolled in a comparative study. The expression of Mfn2 in placenta was detected by qRT-PCR. And the mitochondrial function was detected by ATP assay. Then TEV-1 cells were cultured in hypoxic conditions. mRNA and protein expressions of Mfn2 were detected by qRT-PCR and western blot separately. Cells' viability was detected by MTT. And the mitochondrial function was detected by ATP and mitochondrial membrane potential (MMP) assay. We further knocked down the Mfn2 gene in TEV-1 cells and evaluated the cells' viability. Results. Mfn2 and ATP expressions were significantly decreased in preeclamptic placentae compared to normal placentae. Mfn2 expression level and the viability of TEV-1 cells were reduced during hypoxic conditions. TEV-1 cells' viability, ATP, and MMP levels were also significantly decreased after knockdown of the Mfn2 gene. Conclusions. These results suggest that defects in Mfn2 could cause mitochondrial dysfunction and decrease trophoblastic cells' viability. Therefore, Mfn2 may be functionally involved in the pathogenesis of PE.
Collapse
|
39
|
Abstract
Per milligram of tissue, only the heart exceeds the kidney's abundance of mitochondria. Not surprisingly, renal mitochondria are most densely concentrated in the epithelium of the nephron, at sites where the chemical work of moving solutes against electrochemical gradients places large and constant demands for adenosine triphosphate. Derangements of renal epithelial mitochondria appear to be a hallmark for diverse forms of acute kidney injury (AKI). The pathogenesis of multiple-organ dysfunction syndrome in sepsis is complex, but a substantial body of experimental and observational human data supports the twin concepts that mitochondrial dysfunction contributes to impaired filtration and that recovery of mitochondrial structure and function is essential for recovery from sepsis-associated AKI. These insights have suggested novel methods to diagnose, stratify, prevent, or even treat this common and deadly complication of critical illness. This review will do the following: (1) describe the structure and functions of healthy mitochondria and how renal energy metabolism relates to solute transport; (2) provide an overview of the evidence linking mitochondrial pathology to renal disease; (3) summarize the mitochondrial lesions observed in septic AKI; (4) analyze the role of mitochondrial processes including fission/fusion, mitophagy, and biogenesis in the development of septic AKI and recovery from this disease; and (5) explore the potential for therapeutically targeting mitochondria to prevent or treat septic AKI.
Collapse
|
40
|
Ishimoto Y, Inagi R. Mitochondria: a therapeutic target in acute kidney injury. Nephrol Dial Transplant 2015; 31:1062-9. [DOI: 10.1093/ndt/gfv317] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 08/05/2015] [Indexed: 01/05/2023] Open
|
41
|
Strickland AV, Rebelo AP, Zhang F, Price J, Bolon B, Silva JP, Wen R, Züchner S. Characterization of the mitofusin 2 R94W mutation in a knock-in mouse model. J Peripher Nerv Syst 2015; 19:152-64. [PMID: 24862862 DOI: 10.1111/jns5.12066] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 02/25/2014] [Accepted: 03/19/2014] [Indexed: 11/29/2022]
Abstract
Charcot-Marie-Tooth disease (CMT) comprises a group of heterogeneous peripheral axonopathies affecting 1 in 2,500 individuals. As mutations in several genes cause axonal degeneration in CMT type 2, mutations in mitofusin 2 (MFN2) account for approximately 90% of the most severe cases, making it the most common cause of inherited peripheral axonal degeneration. MFN2 is an integral mitochondrial outer membrane protein that plays a major role in mitochondrial fusion and motility; yet the mechanism by which dominant mutations in this protein lead to neurodegeneration is still not fully understood. Furthermore, future pre-clinical drug trials will be in need of validated rodent models. We have generated a Mfn2 knock-in mouse model expressing Mfn2(R94W), which was originally identified in CMT patients. We have performed behavioral, morphological, and biochemical studies to investigate the consequences of this mutation. Homozygous inheritance leads to premature death at P1, as well as mitochondrial dysfunction, including increased mitochondrial fragmentation in mouse embryonic fibroblasts and decreased ATP levels in newborn brains. Mfn2(R94W) heterozygous mice show histopathology and age-dependent open-field test abnormalities, which support a mild peripheral neuropathy. Although behavior does not mimic the severity of the human disease phenotype, this mouse can provide useful tissues for studying molecular pathways associated with MFN2 point mutations.
Collapse
Affiliation(s)
- Alleene V Strickland
- Department of Human Genetics, Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Iommarini L, Peralta S, Torraco A, Diaz F. Mitochondrial Diseases Part II: Mouse models of OXPHOS deficiencies caused by defects in regulatory factors and other components required for mitochondrial function. Mitochondrion 2015; 22:96-118. [PMID: 25640959 DOI: 10.1016/j.mito.2015.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 01/22/2015] [Indexed: 01/21/2023]
Abstract
Mitochondrial disorders are defined as defects that affect the oxidative phosphorylation system (OXPHOS). They are characterized by a heterogeneous array of clinical presentations due in part to a wide variety of factors required for proper function of the components of the OXPHOS system. There is no cure for these disorders owing to our poor knowledge of the pathogenic mechanisms of disease. To understand the mechanisms of human disease numerous mouse models have been developed in recent years. Here we summarize the features of several mouse models of mitochondrial diseases directly related to those factors affecting mtDNA maintenance, replication, transcription, translation as well as other proteins that are involved in mitochondrial dynamics and quality control which affect mitochondrial OXPHOS function without being intrinsic components of the system. We discuss how these models have contributed to our understanding of mitochondrial diseases and their pathogenic mechanisms.
Collapse
Affiliation(s)
- Luisa Iommarini
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Via Irnerio 42, 40128 Bologna, Italy.
| | - Susana Peralta
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| | - Alessandra Torraco
- Unit for Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Viale di San Paolo, 15 - 00146, Rome, Italy.
| | - Francisca Diaz
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
43
|
Hickey FB, Corcoran JB, Griffin B, Bhreathnach U, Mortiboys H, Reid HM, Andrews D, Byrne S, Furlong F, Martin F, Godson C, Murphy M. IHG-1 increases mitochondrial fusion and bioenergetic function. Diabetes 2014; 63:4314-25. [PMID: 25008184 DOI: 10.2337/db13-1256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Induced in high glucose-1 (IHG-1) is a conserved mitochondrial protein associated with diabetic nephropathy (DN) that amplifies profibrotic transforming growth factor (TGF)-β1 signaling and increases mitochondrial biogenesis. Here we report that inhibition of endogenous IHG-1 expression results in reduced mitochondrial respiratory capacity, ATP production, and mitochondrial fusion. Conversely, overexpression of IHG-1 leads to increased mitochondrial fusion and also protects cells from reactive oxygen species-induced apoptosis. IHG-1 forms complexes with known mediators of mitochondrial fusion-mitofusins (Mfns) 1 and 2-and enhances the GTP-binding capacity of Mfn2, suggesting that IHG-1 acts as a guanine nucleotide exchange factor. IHG-1 must be localized to mitochondria to interact with Mfn1 and Mfn2, and this interaction is necessary for increased IHG-1-mediated mitochondrial fusion. Together, these findings indicate that IHG-1 is a novel regulator of both mitochondrial dynamics and bioenergetic function and contributes to cell survival following oxidant stress. We propose that in diabetic kidney disease increased IHG-1 expression protects cell viability and enhances the actions of TGF-β, leading to renal proximal tubule dedifferentiation, an important event in the pathogenesis of this devastating condition.
Collapse
Affiliation(s)
- Fionnuala B Hickey
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, Ireland Trinity Health Kidney Centre, Trinity College, Dublin, Ireland
| | - James B Corcoran
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, Ireland
| | - Brenda Griffin
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Una Bhreathnach
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, Ireland
| | - Heather Mortiboys
- Academic Neurology Unit, Medical School, University of Sheffield, Sheffield, U.K
| | - Helen M Reid
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Darrell Andrews
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Shane Byrne
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Fiona Furlong
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland
| | - Finian Martin
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, Ireland
| | - Madeline Murphy
- Diabetes Complications Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
44
|
Stacchiotti A, Favero G, Giugno L, Lavazza A, Reiter RJ, Rodella LF, Rezzani R. Mitochondrial and metabolic dysfunction in renal convoluted tubules of obese mice: protective role of melatonin. PLoS One 2014; 9:e111141. [PMID: 25347680 PMCID: PMC4210266 DOI: 10.1371/journal.pone.0111141] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/28/2014] [Indexed: 01/11/2023] Open
Abstract
Obesity is a common and complex health problem, which impacts crucial organs; it is also considered an independent risk factor for chronic kidney disease. Few studies have analyzed the consequence of obesity in the renal proximal convoluted tubules, which are the major tubules involved in reabsorptive processes. For optimal performance of the kidney, energy is primarily provided by mitochondria. Melatonin, an indoleamine and antioxidant, has been identified in mitochondria, and there is considerable evidence regarding its essential role in the prevention of oxidative mitochondrial damage. In this study we evaluated the mechanism(s) of mitochondrial alterations in an animal model of obesity (ob/ob mice) and describe the beneficial effects of melatonin treatment on mitochondrial morphology and dynamics as influenced by mitofusin-2 and the intrinsic apoptotic cascade. Melatonin dissolved in 1% ethanol was added to the drinking water from postnatal week 5–13; the calculated dose of melatonin intake was 100 mg/kg body weight/day. Compared to control mice, obesity-related morphological alterations were apparent in the proximal tubules which contained round mitochondria with irregular, short cristae and cells with elevated apoptotic index. Melatonin supplementation in obese mice changed mitochondria shape and cristae organization of proximal tubules, enhanced mitofusin-2 expression, which in turn modulated the progression of the mitochondria-driven intrinsic apoptotic pathway. These changes possibly aid in reducing renal failure. The melatonin-mediated changes indicate its potential protective use against renal morphological damage and dysfunction associated with obesity and metabolic disease.
Collapse
Affiliation(s)
- Alessandra Stacchiotti
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Lorena Giugno
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Antonio Lavazza
- Istituto Zooprofilattico Sperimentale della Lombardia ed Emilia Romagna, OIE Reference Laboratory for RHD, Brescia, Italy
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health Science Center San Antonio, San Antonio, Texas, United States of America
| | - Luigi Fabrizio Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- * E-mail:
| |
Collapse
|
45
|
Gall JM, Wang Z, Bonegio RG, Havasi A, Liesa M, Vemula P, Borkan SC. Conditional knockout of proximal tubule mitofusin 2 accelerates recovery and improves survival after renal ischemia. J Am Soc Nephrol 2014; 26:1092-102. [PMID: 25201884 DOI: 10.1681/asn.2014010126] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 07/13/2014] [Indexed: 01/23/2023] Open
Abstract
Proximal tubule (PT) cells are critical targets of acute ischemic injury. Elimination of the mitochondrial fusion protein mitofusin 2 (Mfn2) sensitizes PT cells to apoptosis in vitro. However, the role of PT Mfn2 in ischemic AKI in vivo is unknown. To test its role, we evaluated the effects of conditional KO of PT Mfn2 (cKO-PT-Mfn2) on animal survival after transient bilateral renal ischemia associated with severe AKI. Forty-eight hours after ischemia, 28% of control mice survived compared with 86% of cKO-PT-Mfn2 animals (P<0.001 versus control). Although no significant differences in histologic injury score, apoptosis, or necrosis were detected between genotypes, cKO-PT-Mfn2 kidneys exhibited a 3.5-fold increase in cell proliferation restricted to the intrarenal region with Mfn2 deletion. To identify the signals responsible for increased proliferation, primary PT cells with Mfn2 deficiency were subjected to stress by ATP depletion in vitro. Compared with normal Mfn2 expression, Mfn2 deficiency significantly increased PT cell proliferation and persistently activated extracellular signal-regulated kinase 1/2 (ERK1/2) during recovery from stress. Furthermore, stress and Mfn2 deficiency decreased the interaction between Mfn2 and Ras detected by immunoprecipitation, and purified Mfn2 dose-dependently decreased Ras activity in a cell-free assay. Ischemia in vivo also reduced the Mfn2-RAS interaction and increased both RAS and p-ERK1/2 activity in the renal cortical homogenates of cKO-PT-Mfn2 mice. Our results suggest that, in contrast to its proapoptotic effects in vitro, selective PT Mfn2 deficiency accelerates recovery of renal function and enhances animal survival after ischemic AKI in vivo, partly by increasing Ras-ERK-mediated cell proliferation.
Collapse
Affiliation(s)
| | | | | | | | - Marc Liesa
- The Obesity Center, Boston Medical Center, Boston, Massachusetts
| | | | | |
Collapse
|
46
|
Abstract
Acute kidney injury (AKI) is a serious clinical condition with no effective treatment. Tubular cells are key targets in AKI. Tubular cells and, specifically, proximal tubular cells are extremely rich in mitochondria and mitochondrial changes had long been known to be a feature of AKI. However, only recent advances in understanding the molecules involved in mitochondria biogenesis and dynamics and the availability of mitochondria-targeted drugs has allowed the exploration of the specific role of mitochondria in AKI. We now review the morphological and functional mitochondrial changes during AKI, as well as changes in the expression of mitochondrial genes and proteins. Finally, we summarise the current status of novel therapeutic strategies specifically targeting mitochondria such as mitochondrial permeability transition pore (MPTP) opening inhibitors (cyclosporine A (CsA)), quinone analogues (MitoQ, SkQ1 and SkQR1), superoxide dismutase (SOD) mimetics (Mito-CP), Szeto-Schiller (SS) peptides (Bendavia) and mitochondrial division inhibitors (mdivi-1). MitoQ, SkQ1, SkQR1, Mito-CP, Bendavia and mdivi-1 have improved the course of diverse experimental models of AKI. Evidence for a beneficial effect of CsA on human cardiac ischaemia-reperfusion injury derives from a clinical trial; however, CsA is nephrotoxic. MitoQ and Bendavia have been shown to be safe for humans. Ongoing clinical trials are testing the efficacy of Bendavia in AKI prevention following renal artery percutaneous transluminal angioplasty.
Collapse
|
47
|
Zhang L, Yuan X, Wang S, Ou Y, Zheng X, Wang Q. The relationship between mitochondrial fusion/fission and apoptosis in the process of adipose-derived stromal cells differentiation into astrocytes. Neurosci Lett 2014; 575:19-24. [PMID: 24858134 DOI: 10.1016/j.neulet.2014.05.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/11/2014] [Accepted: 05/13/2014] [Indexed: 11/27/2022]
Abstract
To research the features of mitochondrial fusion and fission proteins and their relationship with apoptosis in the process of adipose-derived stromal cells (ADSCs) differentiation into astrocytes. Immunocytochemistry and Western-blotting were used to analyze the expression of glial fibrillary acidic protein (GFAP), mitochondria fusion-2 (Mfn2), mitochondrial fission-1 (Fis1) and cysteine aspartate specific protease-3 (Caspase-3). Flow cytometry for quantification of the number of apoptotic cells. Transmission electron microscopy (TEM) for observation of the ultrastructure. After induced for 48h, 7, 14 and 21 days showed expression of GFAP, reached the peak on the 7th day (P<0.05). The expression of Mfn2 was decreased with the induction time extending, reached the minimum on the 14th day (P<0.05). The expression of Fis1 and Caspase-3 was increased with the induction time extending, reached the peak on the 14th day (P<0.05, respectively). The rates of early apoptosis, late apoptosis or necrosis were increased gradually (P<0.05). Our findings suggest that in the process of ADSCs differentiation into astrocytes, mitochondrial fusion decreased while mitochondrial fission enhanced significantly; caspase-dependent apoptosis was one of the main reasons leading to cell death.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Neurology, Affiliated Kailuan General Hospital of Hebei United University, No. 57, East Xinhua Road, Tangshan 063000, Hebei, China
| | - Xiaodong Yuan
- Department of Neurology, Affiliated Kailuan General Hospital of Hebei United University, No. 57, East Xinhua Road, Tangshan 063000, Hebei, China.
| | - Shujuan Wang
- Department of Neurology, Affiliated Kailuan General Hospital of Hebei United University, No. 57, East Xinhua Road, Tangshan 063000, Hebei, China
| | - Ya Ou
- Department of Neurology, Affiliated Kailuan General Hospital of Hebei United University, No. 57, East Xinhua Road, Tangshan 063000, Hebei, China
| | - Xinyue Zheng
- Department of Neurology, Affiliated Kailuan General Hospital of Hebei United University, No. 57, East Xinhua Road, Tangshan 063000, Hebei, China
| | - Quanquan Wang
- Department of Neurology, Affiliated Kailuan General Hospital of Hebei United University, No. 57, East Xinhua Road, Tangshan 063000, Hebei, China
| |
Collapse
|
48
|
Xiao X, Hu Y, Quirós PM, Wei Q, López-Otín C, Dong Z. OMA1 mediates OPA1 proteolysis and mitochondrial fragmentation in experimental models of ischemic kidney injury. Am J Physiol Renal Physiol 2014; 306:F1318-26. [PMID: 24671334 DOI: 10.1152/ajprenal.00036.2014] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Acute kidney injury (AKI) is associated with mitochondrial fragmentation, which contributes to mitochondrial damage and tubular cell apoptosis. Mitochondrial fragmentation involves the cleavage of both mitochondrial outer and inner membranes. Cleavage of the outer membrane results from Drp-1-mediated fission activation and Bak-promoted fusion arrest, but the molecular mechanism of inner membrane cleavage remains elusive. OMA1-mediated proteolysis of OPA1, a key inner membrane fusion protein, was recently suggested to account for inner membrane cleavage during cell stress. In this study, we determined the role of OMA1 in OPA1 proteolysis and mitochondrial fragmentation in experimental models of ischemic AKI. In ATP-depletion injury, knockdown of OMA1 suppressed OPA1 proteolysis, mitochondrial fragmentation, cytochrome c release, and consequent apoptosis in renal proximal tubular cells. In mice, OMA1 deficiency prevented ischemic AKI as indicated by better renal function, less tubular damage, and lower apoptosis. OPA1 proteolysis and mitochondrial injury during ischemic AKI were ameliorated in OMA1-deficient mice. Thus, OMA1-mediated OPA1 proteolysis plays an important role in the disruption of mitochondrial dynamics in ischemic AKI.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Georgia Reagents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Wuhan University, Wuhan, China; and
| | - Yanzhong Hu
- Department of Cell Biology and Genetics, Henan University School of Medicine, Kaifeng, Henan, China
| | - Pedro M Quirós
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Georgia Reagents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo, Spain
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Georgia Reagents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Cell Biology and Genetics, Henan University School of Medicine, Kaifeng, Henan, China;
| |
Collapse
|
49
|
Che R, Yuan Y, Huang S, Zhang A. Mitochondrial dysfunction in the pathophysiology of renal diseases. Am J Physiol Renal Physiol 2014; 306:F367-78. [PMID: 24305473 DOI: 10.1152/ajprenal.00571.2013] [Citation(s) in RCA: 300] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mitochondrial dysfunction has gained recognition as a contributing factor in many diseases. The kidney is a kind of organ with high energy demand, rich in mitochondria. As such, mitochondrial dysfunction in the kidney plays a critical role in the pathogenesis of kidney diseases. Despite the recognized importance mitochondria play in the pathogenesis of the diseases, there is limited understanding of various aspects of mitochondrial biology. This review examines the physiology and pathophysiology of mitochondria. It begins by discussing mitochondrial structure, mitochondrial DNA, mitochondrial reactive oxygen species production, mitochondrial dynamics, and mitophagy, before turning to inherited mitochondrial cytopathies in kidneys (inherited or sporadic mitochondrial DNA or nuclear DNA mutations in genes that affect mitochondrial function). Glomerular diseases, tubular defects, and other renal diseases are then discussed. Next, acquired mitochondrial dysfunction in kidney diseases is discussed, emphasizing the role of mitochondrial dysfunction in the pathogenesis of chronic kidney disease and acute kidney injury, as their prevalence is increasing. Finally, it summarizes the possible beneficial effects of mitochondrial-targeted therapeutic agents for treatment of mitochondrial dysfunction-mediated kidney injury-genetic therapies, antioxidants, thiazolidinediones, sirtuins, and resveratrol-as mitochondrial-based drugs may offer potential treatments for renal diseases.
Collapse
Affiliation(s)
- Ruochen Che
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Yanggang Yuan
- Department of Nephrology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| | - Aihua Zhang
- Department of Nephrology, Nanjing Children's Hospital, Affiliated with Nanjing Medical University, Nanjing, China
- Institute of Pediatrics, Nanjing Medical University, Nanjing, China; and
| |
Collapse
|
50
|
Stallons LJ, Funk JA, Schnellmann RG. Mitochondrial Homeostasis in Acute Organ Failure. CURRENT PATHOBIOLOGY REPORTS 2013; 1. [PMID: 24386614 DOI: 10.1007/s40139-013-0023-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The kidneys compose approximately 0.5% of the body mass but consume about 10% of the oxygen in cellular respiration. This discordance is due to the high energy demands on the kidney for reabsorption of filtered blood components and makes the kidney sensitive to mitochondrial stress, the primary source of cellular ATP. Regardless of the etiology, acute kidney injury (AKI) almost always involves aspects of mitochondrial dysfunction. Recent evidence from experimental models suggests that preserving mitochondrial function or promoting mitochondrial repair rescues renal function during AKI. In this review we discuss the effect of AKI on disruption of mitochondrial homeostasis, and how the dynamic processes of mitochondrial biogenesis, fission/fusion, and mitophagy influence renal injury and recovery.
Collapse
Affiliation(s)
- L Jay Stallons
- Center for Cell Death, Injury, and Regeneration, Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 280 Calhoun Street, Charleston, South Carolina 29425
| | - Jason A Funk
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, 70 President Street, Charleston, SC 29425
| | - Rick G Schnellmann
- Center for Cell Death, Injury, and Regeneration, Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 280 Calhoun Street, Charleston, South Carolina 29425 ; Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street, Charleston, South Carolina 29401
| |
Collapse
|