1
|
Yang M, Huang Y, Tang A, Zhang Y, Liu Y, Fan Z, Li M. Research Hotspots in Mitochondria-Related Studies for AKI Treatment: A Bibliometric Study. Drug Des Devel Ther 2024; 18:4051-4063. [PMID: 39280255 PMCID: PMC11402358 DOI: 10.2147/dddt.s473426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/27/2024] [Indexed: 09/18/2024] Open
Abstract
Purpose Acute kidney injury (AKI) is a common clinical critical condition that has become a significant healthcare burden. In recent years, the relationship between AKI and mitochondria has attracted increasing attention. Protecting mitochondria or restoring their function has emerged as a novel therapeutic strategy for alleviating AKI. This study aims to analyze and summarize the current status, research trends, and hotspots in this field, providing references and directions for future research. Methods AKI and mitochondria-related literature from the Web of Science core collection were retrieved and collected. Bibliometric and visualization analyses were conducted using Microsoft Excel 2021, bibliometric tools (VosViewer, Citespace 6.3.R1, and the bibliometrix R package), R 4.3.2, and SCImagoGraphica software. Results A total of 2433 publications were included in this study. The number of annual publications in this field has increased year by year. China and the United States are the two most productive countries. Central South University is the most influential research institution in terms of research output, and Parikh SM, Schnellmann RG, and Dong Z are the most influential authors in this field. KIDNEY INTERNATIONAL, JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY, and AMERICAN JOURNAL OF PHYSIOLOGY-RENAL PHYSIOLOGY are the most influential journals. Initially, the research focused on keywords such as oxidative stress, ischemia-reperfusion injury, apoptosis, inflammation, and autophagy. In recent years, new research hotspots have emerged, including ferroptosis, aging, mitochondrial quality control, messenger RNA, mitochondrial-targeted antioxidants, extracellular vesicles, and nanodrug delivery. Conclusion Research on the relationship between mitochondria and AKI has broad developing prospects, and targeting mitochondrial regulation will become a focus of future AKI prevention and treatment research.
Collapse
Affiliation(s)
- Mengfan Yang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Youqun Huang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Anqi Tang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Yu Zhang
- Department of Nephrology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an, Shaanxi Provincial, People’s Republic of China
| | - Yu Liu
- Department of Nephrology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, People’s Republic of China
| | - Zhenliang Fan
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Mingquan Li
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
2
|
Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y, Yang Y. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther 2024; 9:50. [PMID: 38424050 PMCID: PMC10904817 DOI: 10.1038/s41392-024-01756-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family (PGC-1s), consisting of three members encompassing PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC), was discovered more than a quarter-century ago. PGC-1s are essential coordinators of many vital cellular events, including mitochondrial functions, oxidative stress, endoplasmic reticulum homeostasis, and inflammation. Accumulating evidence has shown that PGC-1s are implicated in many diseases, such as cancers, cardiac diseases and cardiovascular diseases, neurological disorders, kidney diseases, motor system diseases, and metabolic disorders. Examining the upstream modulators and co-activated partners of PGC-1s and identifying critical biological events modulated by downstream effectors of PGC-1s contribute to the presentation of the elaborate network of PGC-1s. Furthermore, discussing the correlation between PGC-1s and diseases as well as summarizing the therapy targeting PGC-1s helps make individualized and precise intervention methods. In this review, we summarize basic knowledge regarding the PGC-1s family as well as the molecular regulatory network, discuss the physio-pathological roles of PGC-1s in human diseases, review the application of PGC-1s, including the diagnostic and prognostic value of PGC-1s and several therapies in pre-clinical studies, and suggest several directions for future investigations. This review presents the immense potential of targeting PGC-1s in the treatment of diseases and hopefully facilitates the promotion of PGC-1s as new therapeutic targets.
Collapse
Affiliation(s)
- Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China
| | - Junmin Chen
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China.
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
3
|
Wang T, Huang Y, Zhang X, Zhang Y, Zhang X. Advances in metabolic reprogramming of renal tubular epithelial cells in sepsis-associated acute kidney injury. Front Physiol 2024; 15:1329644. [PMID: 38312312 PMCID: PMC10834781 DOI: 10.3389/fphys.2024.1329644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024] Open
Abstract
Sepsis-associated acute kidney injury presents as a critical condition characterized by prolonged hospital stays, elevated mortality rates, and an increased likelihood of transition to chronic kidney disease. Sepsis-associated acute kidney injury suppresses fatty acid oxidation and oxidative phosphorylation in the mitochondria of renal tubular epithelial cells, thus favoring a metabolic shift towards glycolysis for energy production. This shift acts as a protective mechanism for the kidneys. However, an extended reliance on glycolysis may contribute to tubular atrophy, fibrosis, and subsequent chronic kidney disease progression. Metabolic reprogramming interventions have emerged as prospective strategies to counteract sepsis-associated acute kidney injury by restoring normal metabolic function, offering potential therapeutic and preventive modalities. This review delves into the metabolic alterations of tubular epithelial cells associated with sepsis-associated acute kidney injury, stressing the importance of metabolic reprogramming for the immune response and the urgency of metabolic normalization. We present various intervention targets that could facilitate the recovery of oxidative phosphorylation-centric metabolism. These novel insights and strategies aim to transform the clinical prevention and treatment landscape of sepsis-associated acute kidney injury, with a focus on metabolic mechanisms. This investigation could provide valuable insights for clinicians aiming to enhance patient outcomes in the context of sepsis-associated acute kidney injury.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Critical Care Medicine, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Ying Huang
- Department of Critical Care Medicine, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Xiaobei Zhang
- Department of Critical Care Medicine, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Yi Zhang
- Department of Critical Care Medicine, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Xiangcheng Zhang
- Department of Critical Care Medicine, The Affiliated Huaian No 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| |
Collapse
|
4
|
Jerigova V, Zeman M, Okuliarova M. Chronodisruption of the acute inflammatory response by night lighting in rats. Sci Rep 2023; 13:14109. [PMID: 37644084 PMCID: PMC10465576 DOI: 10.1038/s41598-023-41266-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023] Open
Abstract
Daily oscillations are present in many aspects of the immune system, including responsiveness to infections, allowing temporal alignment of defence mechanisms with the external environment. Our study addresses whether compromised circadian timing function by dim artificial light at night (ALAN) impacts the time dependency of the acute inflammatory response in a rat model of lipopolysaccharide (LPS)-induced inflammation. After 2 weeks of exposure to low-intensity ALAN (~2 lx) or a standard light/dark cycle, male rats were challenged with LPS during either the day or the night. Dim ALAN attenuated the anorectic response when rats were stimulated during their early light phase. Next, ALAN suppressed daily variability in inflammatory changes in blood leukocyte numbers and increased the daytime sensitivity of neutrophils to the priming effects of LPS on oxidative burst. An altered renal inflammatory response in ALAN-exposed rats was manifested by stimulated T-cell infiltration into the kidney upon night-time LPS injection and the modified rhythmic response of genes involved in inflammatory pathways. Moreover, ALAN disturbed steady-state oscillations of the renal molecular clock and eliminated the inflammatory responsiveness of Rev-erbα. Altogether, dim ALAN impaired time-of-day-dependent sensitivity of inflammatory processes, pointing out a causal mechanism between light pollution and negative health effects.
Collapse
Affiliation(s)
- Viera Jerigova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15, Bratislava, Slovakia
| | - Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15, Bratislava, Slovakia
| | - Monika Okuliarova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15, Bratislava, Slovakia.
| |
Collapse
|
5
|
Cuevas-López B, Romero-Ramirez EI, García-Arroyo FE, Tapia E, León-Contreras JC, Silva-Palacios A, Roldán FJ, Campos ONM, Hernandez-Esquivel L, Marín-Hernández A, Gonzaga-Sánchez JG, Hernández-Pando R, Pedraza-Chaverri J, Sánchez-Lozada LG, Aparicio-Trejo OE. NAC Pre-Administration Prevents Cardiac Mitochondrial Bioenergetics, Dynamics, Biogenesis, and Redox Alteration in Folic Acid-AKI-Induced Cardio-Renal Syndrome Type 3. Antioxidants (Basel) 2023; 12:1592. [PMID: 37627587 PMCID: PMC10451243 DOI: 10.3390/antiox12081592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The incidence of kidney disease is increasing worldwide. Acute kidney injury (AKI) can strongly favor cardio-renal syndrome (CRS) type 3 development. However, the mechanism involved in CRS development is not entirely understood. In this sense, mitochondrial impairment in both organs has become a central axis in CRS physiopathology. This study aimed to elucidate the molecular mechanisms associated with cardiac mitochondrial impairment and its role in CRS development in the folic acid-induced AKI (FA-AKI) model. Our results showed that 48 h after FA-AKI, the administration of N-acetyl-cysteine (NAC), a mitochondrial glutathione regulator, prevented the early increase in inflammatory and cell death markers and oxidative stress in the heart. This was associated with the ability of NAC to protect heart mitochondrial bioenergetics, principally oxidative phosphorylation (OXPHOS) and membrane potential, through complex I activity and the preservation of glutathione balance, thus preventing mitochondrial dynamics shifting to fission and the decreases in mitochondrial biogenesis and mass. Our data show, for the first time, that mitochondrial bioenergetics impairment plays a critical role in the mechanism that leads to heart damage. Furthermore, NAC heart mitochondrial preservation during an AKI event can be a valuable strategy to prevent CRS type 3 development.
Collapse
Affiliation(s)
- Belén Cuevas-López
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Edgar Ignacio Romero-Ramirez
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Fernando E. García-Arroyo
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Edilia Tapia
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Juan Carlos León-Contreras
- Experimental Pathology Section, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14000, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - Alejandro Silva-Palacios
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Francisco-Javier Roldán
- Outpatient Department, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Omar Noel Medina Campos
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (O.N.M.C.); (J.P.-C.)
| | - Luz Hernandez-Esquivel
- Department of Biochemistry, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (L.H.-E.); (A.M.-H.)
| | - Alvaro Marín-Hernández
- Department of Biochemistry, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (L.H.-E.); (A.M.-H.)
| | - José Guillermo Gonzaga-Sánchez
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Rogelio Hernández-Pando
- Experimental Pathology Section, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14000, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (O.N.M.C.); (J.P.-C.)
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| |
Collapse
|
6
|
Sharma R, Antypiuk A, Vance SZ, Manwani D, Pearce Q, Cox JE, An X, Yazdanbakhsh K, Vinchi F. Macrophage metabolic rewiring improves heme-suppressed efferocytosis and tissue damage in sickle cell disease. Blood 2023; 141:3091-3108. [PMID: 36952641 PMCID: PMC10315632 DOI: 10.1182/blood.2022018026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 03/16/2023] [Accepted: 03/19/2023] [Indexed: 03/25/2023] Open
Abstract
Sickle cell disease (SCD) is hallmarked by an underlying chronic inflammatory condition, which is contributed by heme-activated proinflammatory macrophages. Although previous studies addressed heme ability to stimulate macrophage inflammatory skewing through Toll-like receptor4 (TLR4)/reactive oxygen species signaling, how heme alters cell functional properties remains unexplored. Macrophage-mediated immune cell recruitment and apoptotic cell (AC) clearance are relevant in the context of SCD, in which tissue damage, cell apoptosis, and inflammation occur owing to vaso-occlusive episodes, hypoxia, and ischemic injury. Here we show that heme strongly alters macrophage functional response to AC damage by exacerbating immune cell recruitment and impairing cell efferocytic capacity. In SCD, heme-driven excessive leukocyte influx and defective efferocytosis contribute to exacerbated tissue damage and sustained inflammation. Mechanistically, these events depend on heme-mediated activation of TLR4 signaling and suppression of the transcription factor proliferator-activated receptor γ (PPARγ) and its coactivator peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α). These changes reduce efferocytic receptor expression and promote mitochondrial remodeling, resulting in a coordinated functional and metabolic reprogramming of macrophages. Overall, this results in limited AC engulfment, impaired metabolic shift to mitochondrial fatty acid β-oxidation, and, ultimately, reduced secretion of the antiinflammatory cytokines interleukin-4 (IL-4) and IL-10, with consequent inhibition of continual efferocytosis, resolution of inflammation, and tissue repair. We further demonstrate that impaired phagocytic capacity is recapitulated by macrophage exposure to plasma of patients with SCD and improved by hemopexin-mediated heme scavenging, PPARγ agonists, or IL-4 exposure through functional and metabolic macrophage rewiring. Our data indicate that therapeutic improvement of heme-altered macrophage functional properties via heme scavenging or PGC1α/PPARγ modulation significantly ameliorates tissue damage associated with SCD pathophysiology.
Collapse
Affiliation(s)
- Richa Sharma
- Iron Research Laboratory, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Ada Antypiuk
- Iron Research Laboratory, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - S. Zebulon Vance
- Iron Research Laboratory, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
| | - Deepa Manwani
- Department of Pediatrics, Albert Einstein College of Medicine, New York, NY
- Pediatric Hematology, The Children's Hospital at Montefiore, New York, NY
| | - Quentinn Pearce
- Department of Biochemistry, University of Utah, Salt Lake City, UT
- Metabolomics, Mass Spectrometry, and Proteomics Core, University of Utah, Salt Lake City, UT
| | - James E. Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT
- Metabolomics, Mass Spectrometry, and Proteomics Core, University of Utah, Salt Lake City, UT
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | | | - Francesca Vinchi
- Iron Research Laboratory, Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
7
|
Tubular Mitochondrial Dysfunction, Oxidative Stress, and Progression of Chronic Kidney Disease. Antioxidants (Basel) 2022; 11:antiox11071356. [PMID: 35883847 PMCID: PMC9311633 DOI: 10.3390/antiox11071356] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected conditions, and CKD is projected to become the fifth leading global cause of death by 2040. New therapeutic approaches are needed. Mitochondrial dysfunction and oxidative stress have emerged as drivers of kidney injury in acute and chronic settings, promoting the AKI-to-CKD transition. In this work, we review the role of mitochondrial dysfunction and oxidative stress in AKI and CKD progression and discuss novel therapeutic approaches. Specifically, evidence for mitochondrial dysfunction in diverse models of AKI (nephrotoxicity, cytokine storm, and ischemia-reperfusion injury) and CKD (diabetic kidney disease, glomerulopathies) is discussed; the clinical implications of novel information on the key role of mitochondria-related transcriptional regulators peroxisome proliferator-activated receptor gamma coactivator 1-alpha, transcription factor EB (PGC-1α, TFEB), and carnitine palmitoyl-transferase 1A (CPT1A) in kidney disease are addressed; the current status of the clinical development of therapeutic approaches targeting mitochondria are updated; and barriers to the clinical development of mitochondria-targeted interventions are discussed, including the lack of clinical diagnostic tests that allow us to categorize the baseline renal mitochondrial dysfunction/mitochondrial oxidative stress and to monitor its response to therapeutic intervention. Finally, key milestones for further research are proposed.
Collapse
|
8
|
Modulation of adipose inflammation by cellular retinoic acid-binding protein 1. Int J Obes (Lond) 2022; 46:1759-1769. [PMID: 35794192 PMCID: PMC9492549 DOI: 10.1038/s41366-022-01175-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/08/2022]
Abstract
Objectives Obesity, a metabolic syndrome, is known to be related to inflammation, especially adipose tissue inflammation. Cellular interactions within the expanded white adipose tissue (WAT) in obesity contribute to inflammation and studies have suggested that inflammation is triggered by inflamed adipocytes that recruit M1 macrophages into WAT. What causes accumulation of unhealthy adipocytes is an important topic of investigation. This study aims to understand the action of Cellular Retinoic Acid Binding Protein 1 (CRABP1) in WAT inflammation. Methods Eight weeks-old wild type (WT) and Crabp1 knockout (CKO) mice were fed with a normal diet (ND) or high-fat diet (HFD) for 8 weeks. Body weight and food intake were monitored. WATs and serum were collected for cellular and molecular analyses to determine affected signaling pathways. In cell culture studies, primary adipocyte differentiation and bone marrow-derived macrophages (BMDM) were used to examine adipocytes’ effects, mediated by CRABP1, in macrophage polarization. The 3T3L1-adipocyte was used to validate relevant signaling pathways. Results CKO mice developed an obese phenotype, more severely under high-fat diet (HFD) feeding. Further, CKO’s WAT exhibited a more severe inflammatory state as compared to wild type (WT) WAT, with a significantly expanded M1-like macrophage population. However, this was not caused by intrinsic defects of CKO macrophages. Rather, CKO adipocytes produced a significantly reduced level of adiponectin and had significantly lowered mitochondrial DNA content. CKO adipocyte-conditioned medium, compared to WT control, inhibited M2-like (CD206+) macrophage polarization. Mechanistically, defects in CKO adipocytes involved the ERK1/2 signaling pathway that could be modulated by CRABP1. Conclusions This study shows that CRABP1 plays a protective role against HFD-induced WAT inflammation through, in part, its regulation of adiponectin production and mitochondrial homeostasis in adipocytes, thereby modulating macrophage polarization in WAT to control its inflammatory potential.
Collapse
|
9
|
Abstract
High-density lipoprotein (HDL) particles, best known for their anti-atherosclerotic effects, also may play a beneficial role during acute renal stress. HDL from healthy human beings also shows anti-inflammatory and anti-oxidant capacities, promotes endothelial function and repair, and serves as a systemic signaling mechanism facilitating rapid interorgan communication during times of physiologic stress. Higher concentrations of HDL are associated with less acute kidney injury after sepsis, cardiac and vascular surgery, and contrast-exposure during percutaneous coronary interventions. A better understanding of the interplay between HDL and the kidney both under homeostatic conditions and under acute physiologic stress could lead to the identification of novel risk factors and therapeutic targets for acute kidney injury prevention and treatment in the future.
Collapse
Affiliation(s)
- Loren E Smith
- Division of Multispecialty Adult Anesthesiology, Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
10
|
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: 245] [Impact Index Per Article: 81.7] [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
|
11
|
Franzin R, Stasi A, Ranieri E, Netti GS, Cantaluppi V, Gesualdo L, Stallone G, Castellano G. Targeting Premature Renal Aging: from Molecular Mechanisms of Cellular Senescence to Senolytic Trials. Front Pharmacol 2021; 12:630419. [PMID: 33995028 PMCID: PMC8117359 DOI: 10.3389/fphar.2021.630419] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/11/2021] [Indexed: 01/10/2023] Open
Abstract
The biological process of renal aging is characterized by progressive structural and functional deterioration of the kidney leading to end-stage renal disease, requiring renal replacement therapy. Since the discovery of pivotal mechanisms of senescence such as cell cycle arrest, apoptosis inhibition, and the development of a senescence-associated secretory phenotype (SASP), efforts in the understanding of how senescent cells participate in renal physiological and pathological aging have grown exponentially. This has been encouraged by both preclinical studies in animal models with senescent cell clearance or genetic depletion as well as due to evidence coming from the clinical oncologic experience. This review considers the molecular mechanism and pathways that trigger premature renal aging from mitochondrial dysfunction, epigenetic modifications to autophagy, DNA damage repair (DDR), and the involvement of extracellular vesicles. We also discuss the different pharmaceutical approaches to selectively target senescent cells (namely, senolytics) or the development of systemic SASP (called senomorphics) in basic models of CKD and clinical trials. Finally, an overview will be provided on the potential opportunities for their use in renal transplantation during ex vivo machine perfusion to improve the quality of the graft.
Collapse
Affiliation(s)
- Rossana Franzin
- Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Alessandra Stasi
- Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Elena Ranieri
- Clinical Pathology, Center of Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Stefano Netti
- Clinical Pathology, Center of Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine and Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Italy
| |
Collapse
|
12
|
Yuan L, Yuan Y, Liu F, Li L, Liu J, Chen Y, Cheng J, Lu Y. PGC-1α alleviates mitochondrial dysfunction via TFEB-mediated autophagy in cisplatin-induced acute kidney injury. Aging (Albany NY) 2021; 13:8421-8439. [PMID: 33714196 PMCID: PMC8034953 DOI: 10.18632/aging.202653] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/19/2020] [Indexed: 02/05/2023]
Abstract
Because of the key role of impaired mitochondria in the progression of acute kidney injury (AKI), it is striking that peroxisome proliferator γ coactivator 1-α (PGC-1α), a transcriptional coactivator of genes involved in mitochondrial biogenesis and autophagy, protects from kidney injury. However, the specific mechanism involved in PGC-1α-mediated autophagy remains elusive. In vivo, along with the severe kidney damage, the expression of PGC-1α was decreased in cisplatin-induced AKI mice. Conversely, PGC-1α activator (ZLN005) administration could alleviate kidney injury. Consistently, in vitro overexpression of PGC-1α or ZLN005 treatment inhibited cell apoptosis and mitochondrial dysfunction induced by cisplatin. Moreover, ZLN005 treatment increased the expression of LC3-II and co-localization between LC3 and mitochondria, suggesting that the mitophagy was activated. Furthermore, PGC-1α-mediated the activation of mitophagy was reliant on the increased expression of TFEB, and the protective effects were abrogated in TFEB-knockdown cells. These data suggest that the activation of PGC-1α could alleviate mitochondrial dysfunction and kidney injury in AKI mice via TFEB-mediated autophagy.
Collapse
Affiliation(s)
- Longhui Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Zhu Y, Xu D, Deng F, Yan Y, Li J, Zhang C, Chu J. Angiotensin (1-7) Attenuates Sepsis-Induced Acute Kidney Injury by Regulating the NF-κB Pathway. Front Pharmacol 2021; 12:601909. [PMID: 33746749 PMCID: PMC7970314 DOI: 10.3389/fphar.2021.601909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
This study explores the protective mechanism of angiotensin (1-7) [Ang-(1-7)] on kidneys by examining its effects on renal histomorphology, inflammatory response, oxidative stress, and NF-κB signaling in mice suffering from sepsis-induced acute kidney injury. A sepsis-induced acute kidney injury mouse model was established by intracervically injecting lipopolysaccharides (LPS group), followed by the administration of Ang-(1-7) [LPS + Ang-(1-7) group]. The serum levels of urea nitrogen, creatinine and cystatin. c were measured with an automatic biochemical analyzer, and changes in proinflammatory cytokines and angiotensin II (Ang II) in the serum and kidneys were quantified by enzyme-linked immunosorbent assays. Changes in oxidative stress indices in the renal cortex were detected by colorimetry. The localization of Ang II in kidneys was examined by immunohistochemistry. Western blotting was used to examine phosphorylated NF-κB-p65 and IκBα levels in kidneys. Compared with the control group, the serum levels of urea nitrogen, creatinine and cystatin. c were increased, whereas the levels of Ang II, TNFα, IL-1β, IL-6, and malondialdehyde (mda) were increased significantly. The levels of Ang II and phosphorylated NF-κB-p65 were elevated in kidneys, whereas the levels of superoxide dismutase (sod), Total antioxidative capacity (TAOC), and inhibitor of NF-κB (IκBα) were reduced in the LPS group (p < 0.05). Pathological damage was also observed in kidneys of LPS-group mice. In Pearson correlation analysis, there was a positive correlation between Ang II and phosphorylated NF-κB-p65 levels, and a negative correlation between Ang II and IκBα levels (p < 0.05). After the application of Ang-(1-7), the levels of urea nitrogen, creatinine, cystatin. c, Ang II, TNFα, IL-1β, IL-6, and mda, as well as the expression of Ang II and phosphorylated NF-κB-p65 in kidneys of LPS + Ang-(1-7)-group mice, were lower than those in kidneys of LPS-group mice, but the levels of sod, TAOC, and IκBα were higher than those of LPS-group mice (p < 0.05). Pathological changes were less severe in mice of the LPS + Ang-(1-7) group. Overall, Ang-(1-7) can decrease the Ang II level, inhibit NF-κB signaling, reduce the inflammatory response, decrease oxidative stress, and mitigate sepsis-associated acute kidney injury.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Nephrology, Anhui Provincial Children's Hospital, Hefei, China
| | - Daliang Xu
- Department of Nephrology, Anhui Provincial Children's Hospital, Hefei, China
| | - Fang Deng
- Department of Nephrology, Anhui Provincial Children's Hospital, Hefei, China
| | - Yonglin Yan
- Department of Nephrology, Anhui Provincial Children's Hospital, Hefei, China
| | - Jian Li
- Department of Nephrology, Anhui Provincial Children's Hospital, Hefei, China
| | - Chenyu Zhang
- Department of Clinical Laboratory, Anhui Provincial Children's Hospital, Hefei, China
| | - Jing Chu
- Department of Pathology, Anhui Provincial Children's Hospital, Hefei, China
| |
Collapse
|
14
|
Veres B, Eros K, Antus C, Kalman N, Fonai F, Jakus PB, Boros E, Hegedus Z, Nagy I, Tretter L, Gallyas F, Sumegi B. Cyclophilin D-dependent mitochondrial permeability transition amplifies inflammatory reprogramming in endotoxemia. FEBS Open Bio 2021; 11:684-704. [PMID: 33471430 PMCID: PMC7931201 DOI: 10.1002/2211-5463.13091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/16/2020] [Accepted: 12/30/2020] [Indexed: 01/05/2023] Open
Abstract
Microorganisms or LPS (lipopolysaccharide), an outer membrane component of Gram-negative bacteria, can induce a systemic inflammatory response that leads to sepsis, multiple organ dysfunction, and mortality. Here, we investigated the role of cyclophilin D (CypD)-dependent mitochondrial permeability transition (mPT) in the immunosuppressive phase of LPS-induced endotoxic shock. The liver plays an important role in immunity and organ dysfunction; therefore, we used liver RNA sequencing (RNA-seq) data, Ingenuity® Pathway Analysis (IPA ® ) to investigate the complex role of mPT formation in inflammatory reprogramming and disease progression. LPS induced significant changes in the expression of 2844 genes, affecting 179 pathways related to mitochondrial dysfunction, defective oxidative phosphorylation, nitric oxide (NO) and reactive oxygen species (ROS) accumulation, nuclear factor, erythroid 2 like 2 (Nrf2), Toll-like receptors (TLRs), and tumor necrosis factor α receptor (TNFR)-mediated processes in wild-type mice. The disruption of CypD reduced LPS-induced alterations in gene expression and pathways involving TNFRs and TLRs, in addition to improving survival and attenuating oxidative liver damage and the related NO- and ROS-producing pathways. CypD deficiency diminished the suppressive effect of LPS on mitochondrial function, nuclear- and mitochondrial-encoded genes, and mitochondrial DNA (mtDNA) quantity, which could be critical in improving survival. Our data propose that CypD-dependent mPT is an amplifier in inflammatory reprogramming and promotes disease progression. The mortality in human sepsis and shock is associated with mitochondrial dysfunction. Prevention of mPT by CypD disruption reduces inflammatory reprogramming, mitochondrial dysfunction, and lethality; therefore, CypD can be a novel drug target in endotoxic shock and related inflammatory diseases.
Collapse
Affiliation(s)
- Balazs Veres
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Krisztian Eros
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
- MTA‐PTE Nuclear‐Mitochondrial Interactions Research GroupPecsHungary
- Szentagothai Janos Research CenterUniversity of PecsHungary
| | - Csenge Antus
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Nikoletta Kalman
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Fruzsina Fonai
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Peter Balazs Jakus
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
| | - Eva Boros
- Institute of BiochemistryBiological Research CentreSzegedHungary
| | - Zoltan Hegedus
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
- Institute of BiophysicsBiological Research CentreSzegedHungary
| | - Istvan Nagy
- Institute of BiochemistryBiological Research CentreSzegedHungary
- SeqOmics Biotechnology LtdMorahalomHungary
| | - Laszlo Tretter
- Department of Medical BiochemistrySemmelweis UniversityBudapestHungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
- MTA‐PTE Nuclear‐Mitochondrial Interactions Research GroupPecsHungary
- Szentagothai Janos Research CenterUniversity of PecsHungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical ChemistryMedical SchoolUniversity of PecsHungary
- MTA‐PTE Nuclear‐Mitochondrial Interactions Research GroupPecsHungary
- Szentagothai Janos Research CenterUniversity of PecsHungary
| |
Collapse
|
15
|
Serum Mitochondrial Quality Control Related Biomarker Levels are Associated with Organ Dysfunction in Septic Patients. Shock 2021; 56:412-418. [PMID: 33534397 DOI: 10.1097/shk.0000000000001737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND To investigate the feasibility and the value of using mitochondrial quality control (MQC)-related proteins as biomarkers in septic patients. METHODS The enrolled subjects were divided into four groups: healthy control group (n = 30), intensive care unit (ICU) control group (n = 62), septic nonshock group (n = 40), and septic shock group (n = 94). Serum levels of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), fission protein 1 (Fis1), mitofusin2 (Mfn2), and Parkin were measured by enzyme-linked immunosorbent assay at the time of enrollment for all groups. Clinical parameters and laboratory test results were also collected. RESULTS The levels of MQC-related biomarkers between any two of the four groups were significantly different (P < 0.001 for all). The serum levels of PGC-1α, Mfn2, and Parkin were lowest in healthy individuals; the levels were dramatically higher in the ICU control group compared with the others, and they decreased progressively from the septic nonshock group to the septic shock group. However, the pattern for Fis1 was inverse; the more severe the condition was, the higher the level of Fis1. Moreover, there was moderate correlation between MQC-related biomarkers and the SOFA score (PGC-1α, r = -0.662; Fis1, r = 0.609; Mfn2, r = -0.677; Parkin, r = 0.-0.674, P < 0.001 for all). CONCLUSIONS The serum levels of PGC-1α, Fis1, Mfn2, and Parkin were significantly correlated with organ dysfunction and reflected the disease progression and severity. The dynamic surveillance of these four biomarkers could be beneficial to predict outcome and guide treatment.
Collapse
|
16
|
Tubular mitochondrial AKT1 is activated during ischemia reperfusion injury and has a critical role in predisposition to chronic kidney disease. Kidney Int 2020; 99:870-884. [PMID: 33316281 DOI: 10.1016/j.kint.2020.10.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 12/21/2022]
Abstract
Kidney tubular dysfunction contributes to acute kidney injury and to the transition to chronic kidney disease. Although tubular mitochondria have been implicated in the pathophysiology of kidney failure, the mechanisms are not yet clear. Here, we demonstrated that ischemia-reperfusion injury induced acute translocation and activation of mitochondrial protein kinase B (also known as AKT1) in the kidney tubules. We hypothesized that mitochondrial AKT1 signaling protects against the development of acute kidney injury and subsequent chronic kidney disease. To test this prediction, we generated two novel kidney tubule-specific transgenic mouse strains with inducible expression of mitochondria-targeted dominant negative AKT1 or constitutively active AKT1, using a Cre-Lox strategy. Inhibition of mitochondrial AKT1 in mitochondria-targeted dominant negative AKT1 mice aggravated azotemia, tubular injuries, kidney fibrosis, glomerulosclerosis, and negatively impacted survival after ischemia-reperfusion injury. Conversely, enhancing tubular mitochondrial AKT1 signaling in mitochondria-targeted constitutively active AKT1 mice attenuated kidney injuries, protected kidney function, and significantly improved survival after ischemia-reperfusion injury (76.9% vs. 20.8%, respectively). Uncoupled mitochondrial respiration and increased oxidative stress was found in the kidney tubules when mitochondria AKT1 was inhibited, supporting the role of mitochondrial dysfunction in the pathophysiology of kidney failure. Thus, our studies suggest tubular mitochondrial AKT1 signaling could be a novel target to develop new strategies for better prevention and treatment of kidney injury.
Collapse
|
17
|
Clark AJ, Parikh SM. Targeting energy pathways in kidney disease: the roles of sirtuins, AMPK, and PGC1α. Kidney Int 2020; 99:828-840. [PMID: 33307105 DOI: 10.1016/j.kint.2020.09.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 12/16/2022]
Abstract
The kidney has extraordinary metabolic demands to sustain the active transport of solutes that is critical to renal filtration and clearance. Mitochondrial health is vital to meet those demands and maintain renal fitness. Decades of studies have linked poor mitochondrial health to kidney disease. Key regulators of mitochondrial health-adenosine monophosphate kinase, sirtuins, and peroxisome proliferator-activated receptor γ coactivator-1α-have all been shown to play significant roles in renal resilience against disease. This review will summarize the latest research into the activities of those regulators and evaluate the roles and therapeutic potential of targeting those regulators in acute kidney injury, glomerular kidney disease, and renal fibrosis.
Collapse
Affiliation(s)
- Amanda J Clark
- Division of Nephrology, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Samir M Parikh
- Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
| |
Collapse
|
18
|
Nakatomi K, Ueno H, Ishikawa Y, Salim RC, Mori Y, Kanemoto I, Tancharoen S, Kikuchi K, Miura N, Omori T, Okuda-Ashitaka E, Matsumura K, Imaizumi H, Motomiya Y, Maruyama I, Kawahara KI. TLR4/MD-2 is a receptor for extracellular nucleophosmin 1. Biomed Rep 2020; 14:21. [PMID: 33335727 PMCID: PMC7739869 DOI: 10.3892/br.2020.1397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
Nucleophosmin 1 (NPM1) primarily localizes to the nucleus and is passively released into the extracellular milieu by necrotic or damaged cells, or is secreted by monocytes and macrophages. Extracellular NPM1 acts as a potent inflammatory stimulator by promoting cytokine production [e.g., tumor necrosis factor-α (TNF-α)], which suggests that NPM1 acts as a damage-associated molecular pattern. However, the receptor of NPM1 is unknown. Evidence indicates that DAMPs, which include high mobility group box 1 and histones, may bind Toll-like receptors (TLRs). In the present study, it was shown that NPM1 signaling was mediated via the TLR4 pathway, which suggests that TLR4 is an NPM1 receptor. TLR4 binds myeloid differentiation protein-2 (MD-2), which is essential for intracellular signaling. Furthermore, the TLR4 antagonist, LPS-Rhodobacter sphaeroides (an MD-2 antagonist) and TAK-242 (a TLR4 signaling inhibitor) significantly inhibited NPM1-induced TNF-α production by differentiated THP-1 cells as well as reducing ERK1/2 activation. Far-western blot analysis revealed that NPM1 directly bound MD-2. Thus, the results of the present study provide compelling evidence that TLR4 binds NPM1, and it is hypothesized that inhibiting NPM1 activity may serve as a novel strategy for treating TLR4-related diseases.
Collapse
Affiliation(s)
- Kota Nakatomi
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Hikari Ueno
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Yuto Ishikawa
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Ronny Christiadi Salim
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Yuki Mori
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Issey Kanemoto
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Salunya Tancharoen
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan.,Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Kiyoshi Kikuchi
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand.,Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan.,Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8520, Japan
| | - Naoki Miura
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-8580, Japan
| | - Taketo Omori
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Emiko Okuda-Ashitaka
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Kiyoshi Matsumura
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Hitoshi Imaizumi
- Department of Anesthesiology and Intensive Care Medicine, Tokyo Medical University, School of Medicine, Tokyo 160-0023, Japan
| | | | - Ikuro Maruyama
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan.,Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8520, Japan
| | - Ko-Ichi Kawahara
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan.,Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8520, Japan
| |
Collapse
|
19
|
Zhou L, Huntington K, Zhang S, Carlsen L, So EY, Parker C, Sahin I, Safran H, Kamle S, Lee CM, Geun Lee C, A. Elias J, S. Campbell K, T. Naik M, J. Atwood W, Youssef E, A. Pachter J, Navaraj A, A. Seyhan A, Liang O, El-Deiry WS. MEK inhibitors reduce cellular expression of ACE2, pERK, pRb while stimulating NK-mediated cytotoxicity and attenuating inflammatory cytokines relevant to SARS-CoV-2 infection. Oncotarget 2020; 11:4201-4223. [PMID: 33245731 PMCID: PMC7679035 DOI: 10.18632/oncotarget.27799] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/17/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19 affects vulnerable populations including elderly individuals and patients with cancer. Natural Killer (NK) cells and innate-immune TRAIL suppress transformed and virally-infected cells. ACE2, and TMPRSS2 protease promote SARS-CoV-2 infectivity, while inflammatory cytokines IL-6, or G-CSF worsen COVID-19 severity. We show MEK inhibitors (MEKi) VS-6766, trametinib and selumetinib reduce ACE2 expression in human cells. In some human cells, remdesivir increases ACE2-promoter luciferase-reporter expression, ACE2 mRNA and protein, and ACE2 expression is attenuated by MEKi. In serum-deprived and stimulated cells treated with remdesivir and MEKi we observed correlations between pRB, pERK, and ACE2 expression further supporting role of proliferative state and MAPK pathway in ACE2 regulation. We show elevated cytokines in COVID-19-(+) patient plasma (N = 9) versus control (N = 11). TMPRSS2, inflammatory cytokines G-CSF, M-CSF, IL-1α, IL-6 and MCP-1 are suppressed by MEKi alone or with remdesivir. We observed MEKi stimulation of NK-cell killing of target-cells, without suppressing TRAIL-mediated cytotoxicity. Pseudotyped SARS-CoV-2 virus with a lentiviral core and SARS-CoV-2 D614 or G614 SPIKE (S) protein on its envelope infected human bronchial epithelial cells, small airway epithelial cells, or lung cancer cells and MEKi suppressed infectivity of the pseudovirus. We show a drug class-effect with MEKi to stimulate NK cells, inhibit inflammatory cytokines and block host-factors for SARS-CoV-2 infection leading also to suppression of SARS-CoV-2-S pseudovirus infection of human cells. MEKi may attenuate SARS-CoV-2 infection to allow immune responses and antiviral agents to control disease progression.
Collapse
Affiliation(s)
- Lanlan Zhou
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- These authors contributed equally to this work
| | - Kelsey Huntington
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- These authors contributed equally to this work
| | - Shengliang Zhang
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Lindsey Carlsen
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Eui-Young So
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Cassandra Parker
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Surgery, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Ilyas Sahin
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Howard Safran
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Suchitra Kamle
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Chang-Min Lee
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Chun Geun Lee
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Jack A. Elias
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Kerry S. Campbell
- Blood Cell and Development Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Mandar T. Naik
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Molecular Pharmacology, Physiology and Biotechnology, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Walter J. Atwood
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | | | | | - Arunasalam Navaraj
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Attila A. Seyhan
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Olin Liang
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Wafik S. El-Deiry
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI 02912, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI 02912, USA
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Pathobiology Graduate Program, Brown University, Providence, RI 02912, USA
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| |
Collapse
|
20
|
Zhong Y, Wu S, Yang Y, Li GQ, Meng L, Zheng QY, Li Y, Xu GL, Zhang KQ, Peng KF. LIGHT aggravates sepsis-associated acute kidney injury via TLR4-MyD88-NF-κB pathway. J Cell Mol Med 2020; 24:11936-11948. [PMID: 32881263 PMCID: PMC7579683 DOI: 10.1111/jcmm.15815] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/30/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Sepsis‐associated acute kidney injury (SA‐AKI) is a common clinical critical care syndrome. It has received increasing attention due to its high morbidity and mortality; however, its pathophysiological mechanisms remain elusive. LIGHT, the 14th member of the tumour necrosis factor (TNF) superfamily and a bidirectional immunoregulatory molecule that regulates inflammation, plays a pivotal role in disease pathogenesis. In this study, mice with an intraperitoneal injection of LPS and HK‐2 cells challenged with LPS were employed as a model of SA‐AKI in vivo and in vitro, respectively. LIGHT deficiency notably attenuated kidney injury in pathological damage and renal function and markedly mitigated the inflammatory reaction by decreasing inflammatory mediator production and inflammatory cell infiltration in vivo. The TLR4‐Myd88‐NF‐κB signalling pathway in the kidney of LIGHT knockout mice was dramatically down‐regulated compared to the controls. Recombinant human LIGHT aggravated LPS‐treated HK‐2 cell injury by up‐regulating the expression of the TLR4‐Myd88‐NF‐κB signalling pathway and inflammation levels. TAK 242 (a selective TLR4 inhibitor) reduced this trend to some extent. In addition, blocking LIGHT with soluble receptor fusion proteins HVEM‐Fc or LTβR‐Fc in mice attenuated renal dysfunction and pathological damage in SA‐AKI. Our findings indicate that LIGHT aggravates inflammation and promotes kidney damage in LPS‐induced SA‐AKI via the TLR4‐Myd88‐NF‐κB signalling pathway, which provide potential strategies for the treatment of SA‐AKI.
Collapse
Affiliation(s)
- Yu Zhong
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Shun Wu
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yan Yang
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Gui-Qing Li
- Department of Immunology, Army Medical University, Chongqing, China
| | - Li Meng
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Quan-You Zheng
- Department of Urology, 958 Hospital, Southwest Hospital, Army Medical University, Chongqing, China
| | - You Li
- Department of intense care, Daping Hospital, Army Medical University, Chongqing, China
| | - Gui-Lian Xu
- Department of Immunology, Army Medical University, Chongqing, China
| | - Ke-Qin Zhang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kan-Fu Peng
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
21
|
Zhou L, Huntington K, Zhang S, Carlsen L, So EY, Parker C, Sahin I, Safran H, Kamle S, Lee CM, Lee CG, Elias JA, Campbell KS, Naik MT, Atwood WJ, Youssef E, Pachter JA, Navaraj A, Seyhan AA, Liang O, El-Deiry WS. Natural Killer cell activation, reduced ACE2, TMPRSS2, cytokines G-CSF, M-CSF and SARS-CoV-2-S pseudovirus infectivity by MEK inhibitor treatment of human cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.08.02.230839. [PMID: 32793908 PMCID: PMC7418728 DOI: 10.1101/2020.08.02.230839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
COVID-19 affects vulnerable populations including elderly individuals and patients with cancer. Natural Killer (NK) cells and innate-immune TRAIL suppress transformed and virally-infected cells. ACE2, and TMPRSS2 protease promote SARS-CoV-2 infectivity, while inflammatory cytokines IL-6, or G-CSF worsen COVID-19 severity. We show MEK inhibitors (MEKi) VS-6766, trametinib and selumetinib reduce ACE2 expression in human cells. In some human cells, remdesivir increases ACE2-promoter luciferase-reporter expression, ACE2 mRNA and protein, and ACE2 expression is attenuated by MEKi. In serum-deprived and stimulated cells treated with remdesivir and MEKi we observed correlations between pRB, pERK, and ACE2 expression further supporting role of proliferative state and MAPK pathway in ACE2 regulation. We show elevated cytokines in COVID-19-(+) patient plasma (N=9) versus control (N=11). TMPRSS2, inflammatory cytokines G-CSF, M-CSF, IL-1α, IL-6 and MCP-1 are suppressed by MEKi alone or with remdesivir. We observed MEKi stimulation of NK-cell killing of target-cells, without suppressing TRAIL-mediated cytotoxicity. Pseudotyped SARS-CoV-2 virus with a lentiviral core and SARS-CoV-2 D614 or G614 SPIKE (S) protein on its envelope infected human bronchial epithelial cells, small airway epithelial cells, or lung cancer cells and MEKi suppressed infectivity of the pseudovirus. We show a drug class-effect with MEKi to stimulate NK cells, inhibit inflammatory cytokines and block host-factors for SARS-CoV-2 infection leading also to suppression of SARS-CoV-2-S pseudovirus infection of human cells. MEKi may attenuate SARS-CoV-2 infection to allow immune responses and antiviral agents to control disease progression.
Collapse
Affiliation(s)
- Lanlan Zhou
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Kelsey Huntington
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Pathobiology Graduate Program, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Shengliang Zhang
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Lindsey Carlsen
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Pathobiology Graduate Program, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Eui-Young So
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Cassandra Parker
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Surgery, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Ilyas Sahin
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Howard Safran
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Suchitra Kamle
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Chang-Min Lee
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Chun Geun Lee
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Jack A. Elias
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Kerry S. Campbell
- Blood Cell and Development Program, Fox Chase Cancer Center, Philadelphia, PA, 19111
| | - Mandar T. Naik
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Molecular Pharmacology, Physiology and Biotechnology, Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Walter J. Atwood
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Molecular Biology, Cell Biology, and Biochemistry, Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | | | | | - Arunasalam Navaraj
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Attila A. Seyhan
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Olin Liang
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
| | - Wafik S. El-Deiry
- Brown Experimentalists Against COVID-19 (BEACON) Group, Brown University, Providence, RI, 02912
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, RI, 02912
- Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Pathobiology Graduate Program, Brown University, Providence, RI, 02912
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Warren Alpert Medical School, Brown University, Providence, RI, 02912
- Correspondence:
| |
Collapse
|
22
|
Collier JB, Schnellmann RG. Extracellular signal-regulated kinase 1/2 regulates NAD metabolism during acute kidney injury through microRNA-34a-mediated NAMPT expression. Cell Mol Life Sci 2020; 77:3643-3655. [PMID: 31873757 PMCID: PMC11104937 DOI: 10.1007/s00018-019-03391-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 10/26/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
Prior studies have established the important role of extracellular signal-regulated kinase 1/2 (ERK1/2) as a mediator of acute kidney injury (AKI). We demonstrated rapid ERK1/2 activation induced renal dysfunction following ischemia/reperfusion (IR)-induced AKI and downregulated the mitochondrial biogenesis (MB) regulator, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) in mice. In this study, ERK1/2 regulation of cellular nicotinamide adenine dinucleotide (NAD) and PGC-1α were explored. Inhibition of ERK1/2 activation during AKI in mice using the MEK1/2 inhibitor, trametinib, attenuated renal cortical oxidized NAD (NAD+) depletion. The rate-limiting NAD biosynthesis salvage enzyme, NAMPT, decreased following AKI, and this decrease was prevented by ERK1/2 inhibition. The microRNA miR34a decreased with the inhibition of ERK1/2, leading to increased NAMPT protein. Mice treated with a miR34a mimic prevented increases in NAMPT protein in the renal cortex in the presence of ERK1/2 inhibition. In addition, ERK1/2 activation increased acetylated PGC-1α, the less active form, whereas inhibition of ERK1/2 activation prevented an increase in acetylated PGC-1α after AKI through SIRT1 and NAD+ attenuation. These results implicate IR-induced ERK1/2 activation as an important contributor to the downregulation of both PGC-1α and NAD+ pathways that ultimately decrease cellular metabolism and renal function. Inhibition of ERK1/2 activation prior to the initiation of IR injury attenuated decreases in PGC-1α and NAD+ and prevented kidney dysfunction.
Collapse
Affiliation(s)
- Justin B Collier
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA.
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA.
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA.
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
- Southern Arizona VA Health Care System, Tucson, AZ, USA
- Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
23
|
Lu J, Zhang L, Cheng L, He S, Zhang Y, Yan J, Zhou J. Xijiao Dihuang decoction improves prognosis of sepsis via inhibition of aerobic glycolysis. Biomed Pharmacother 2020; 129:110501. [PMID: 32768976 DOI: 10.1016/j.biopha.2020.110501] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Aerobic glycolysis is a key factor to aggravate progression of sepsis. Xijiao Dihuang decoction (XJDHT) has been proven to have favorable therapeutic effects on sepsis. Our previous study has shown that XJDHT is capable of improving survival from sepsis. In this study we investigated the effects of XJDHT on aerobic glycolysis. The rats were randomly divided into five groups, which included control group, model group, TAK-242 group, XJDHT (25 g/kg) group and XJDHT (12.5 g/kg) group. The contents of cytokines increased in the model group compared with control group, while XJDHT reduced expressions of cytokines. Furthermore, the expressions of TLR4, HIF-1α and PKM2 were reduced significantly in the XJDHT group compared with the model group. There were five groups, including control group, LPS group, siTLR4 group, XJDHT (4 mg/mL) group and XJDHT (2 mg/mL) group in vitro experiments. The IL-1β and IL-6 were elevated significantly after LPS stimulation in the model group, while XJDHT reduced the expression of cytokines. Protein expressions of TLR4, HIF-1α and PKM2 were increased significantly by stimulation of LPS, while XJDHT down-regulated the expressions of key molecules in the signaling pathway. To conclude, our study implies that XJDHT is capable of improving the prognosis of sepsis by inhibiting aerobic glycolysis via down-regulation of TLR4/HIF-1α/PKM2 signaling pathway.
Collapse
Affiliation(s)
- Jun Lu
- Department of Intensive Care Unit, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Luyao Zhang
- Department of Pathology, Nanjing University of Chinese Medicine, Nanjing 210013, China
| | - Lu Cheng
- Department of Intensive Care Unit, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Shuyin He
- Department of Intensive Care Unit, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Yan Zhang
- Department of Intensive Care Unit, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Jing Yan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210013, China.
| | - Jiang Zhou
- Department of Intensive Care Unit, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
24
|
Empagliflozin suppresses inflammation and protects against acute septic renal injury. Inflammopharmacology 2020; 29:269-279. [PMID: 32564182 DOI: 10.1007/s10787-020-00732-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sepsis-induced systemic inflammation response syndrome is the leading cause of morbidity and mortality among patients in intensive care units in North America. While sepsis is associated with multiple organ damage, acute renal injury represents a hallmark of sepsis. Since systemic and renal inflammation is known to play a vital role in morbidity and mortality associated with sepsis, identifying a potent anti-inflammatory agent may help minimize morbidity and mortality associated with acute septic kidney injury. Since recent work has suggested that empagliflozin, a renal sodium-glucose cotransporter 2 (SGLT2) inhibitor, may assist in the treatment of inflammatory diseases, our objective was to examine the effect of empagliflozin on acute sepsis-induced renal injury. METHOD Mice were treated with three daily doses of empagliflozin or vehicle, with lipopolysaccharide (LPS) administered on the third day, at the same time as the third dose of empagliflozin or vehicle. In another cohort, mice were injected with a single dose of LPS 3 h before a dose of empagliflozin. RESULTS Our results show that empagliflozin improves survival in a mouse model of LPS-induced septic shock. We further demonstrate that the beneficial effects of empagliflozin are likely mediated via reducing LPS-induced acute renal injury. Moreover, our data indicate that empagliflozin significantly reduces systemic and renal inflammation to contribute to the improvements observed in an LPS-model of acute septic renal injury. CONCLUSION Overall, the findings of this study suggest that empagliflozin could be repurposed to reduce morbidity and mortality in patients with acute septic renal injury. TRIAL REGISTRATION Not applicable.
Collapse
|
25
|
Zhang C, Jia Y, Liu B, Wang G, Zhang Y. TLR4 knockout upregulates the expression of Mfn2 and PGC-1α in a high-fat diet and ischemia-reperfusion mice model of liver injury. Life Sci 2020; 254:117762. [PMID: 32437795 DOI: 10.1016/j.lfs.2020.117762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 12/30/2022]
Abstract
AIMS Patients with nonalcoholic fatty liver disease (NAFLD) have less tolerance to ischemia-reperfusion injury (IRI) of the liver than those with the healthy liver; hence have a higher incidence of severe complications after surgery. This study aimed to investigate the dynamics of the liver and mitochondrial damage and the impact of TLR4 knockout (TLR4KO) on Mfn2 expression in the composite model of NAFLD and IRI. MAIN METHODS We performed high-fat diet (HFD) feeding and ischemia reperfusion (IR) on wild type (WT) and TLR4 knockout TLR4KO mice. KEY FINDINGS The degree of structural and functional injuries to the liver and mitochondria (NAFLD and IRI) is greater than that caused by a single factor (NAFLD or IRI) or a simple superposition of both. The IL-6 and TNF-α expressions were significantly suppressed (P < .05), while PGC-1α and Mfn2 expressions were up-regulated considerably (P < .05) after TLR4KO. Furthermore, mitochondrial fusion increased, while ATP consumption and ROS production decreased significantly after TLR4KO (P < .05). The degree of reduction of compound injury by TLR4KO is more significant than the reduction degree of single factor injury. Also, TNF-α and IL-6 levels can be used predictive markers for mitochondrial damage and liver tolerance to NAFLD and IRI. SIGNIFICANCE TLR4KO upregulates the expression of Mfn2 and PGC-1α in the composite model of NAFLD and IRI. This pathway may be related to IL-6 and TNF-α. This evidence provides theoretical and experimental basis for the subsequent Toll-like receptor 4 (TLR4) receptor targeted therapy.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yinzhao Jia
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
26
|
JNK Signaling Pathway Suppresses LPS-Mediated Apoptosis of HK-2 Cells by Upregulating NGAL. Int J Inflam 2020; 2020:3980507. [PMID: 32373311 PMCID: PMC7196152 DOI: 10.1155/2020/3980507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/19/2020] [Accepted: 04/08/2020] [Indexed: 11/28/2022] Open
Abstract
Objective To explore the role of the c-Jun N-terminal kinase (JNK) signaling pathway in upregulated NGAL expression and its antiapoptotic mechanism in lipopolysaccharide (LPS)-mediated renal tubular epithelial cell injury. Methods In vitro, HK-2 cells were divided into five groups (Con, LPS 1 h, LPS 3 h, LPS 6 h, and LPS 12 h groups) based on the time of LPS (10 μM) treatment. NGAL and caspase-3 gene expression levels were detected by RT-PCR to assess dynamic changes. HK-2 cells were pretreated with SP600125 (20 μM) for 2 hours, followed by LPS (10 μM) stimulation for 3 hours. NGAL and caspase-3 gene expression levels were then determined. Results NGAL mRNA was increased significantly within 6 hours, and caspase-3 mRNA was increased within 3 hours after treatment (P < 0.05). Correlation analysis showed a high correlation between their expression (r = 0.448, P < 0.05). After pretreatment with SP600125, mRNA expression of NGAL in the LPS group was inhibited, while that of caspase-3 was increased significantly. The NGAL mRNA expression level in the SB + LPS group was decreased significantly compared with that in the LPS group, but it was slightly higher than that in the SP group (∼1.5 times of that in the Con group). However, caspase-3 mRNA expression was increased significantly in the SB + LPS group (P < 0.001) (3.5 times of that in the Con group). It also showed a significant increase compared with SP and LPS groups (P < 0.001 vs. SB group; P < 0.05 vs. LPS group). We also found that NGAL and caspase 3 proteins were increased significantly in LPS and SP + LPS groups, but SP600125 decreased the NGAL level by almost 35% and increased the caspase 3 level by 50% in the SP + LPS group compared with the LPS group (P < 0.05). Conclusions The JNK signaling pathway inhibits LPS-mediated apoptosis of renal tubular epithelial cells by upregulating NGAL.
Collapse
|
27
|
Brown CN, Atwood DJ, Pokhrel D, Ravichandran K, Holditch SJ, Saxena S, Miyazaki M, Nemenoff R, Weiser-Evans MCM, Ljubanovic DG, Joy MS, Edelstein CL. The effect of MEK1/2 inhibitors on cisplatin-induced acute kidney injury (AKI) and cancer growth in mice. Cell Signal 2020; 71:109605. [PMID: 32194168 DOI: 10.1016/j.cellsig.2020.109605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/13/2020] [Accepted: 03/15/2020] [Indexed: 12/11/2022]
Abstract
In a clinically-relevant model of 4 week, low-dose cisplatin-induced AKI, mice were injected subcutaneously with non small cell lung cancer (NSCLC) cells that harbor an activating Kirsten rat sarcoma viral oncogene homolog (KRAS)G12V mutation. Phospho extracellular signal-regulated kinase1/2 (pERK1/2) expression in kidney and tumors was decreased by the MEK1/2 inhibitors, U0126 and trametinib, that potently inhibit pERK1/2. U0126 resulted in a significant improvement in kidney function, acute tubular necrosis (ATN) and tubular cell apoptosis in mice with AKI. Genes that were significantly decreased by U0126 were heat shock protein 1, cyclin-dependent kinase 4 (CDK4) and stratifin (14-3-3σ). U0126 resulted in a significant decrease in tumor weight and volume and significantly increased the chemotherapeutic effect of cisplatin. Trametinib, a MEK1/2 inhibitor that is FDA-approved for the treatment of cancer, did not result in functional protection against AKI or worse AKI, but dramatically decreased tumor growth more than cisplatin. Smaller tumors in cisplatin or MEK1/2 inhibitor-treated mice were not related to changes in microtubule-associated proteins 1A/1B light chain 3B (LC3-II), p62, cleaved caspase-3, granzyme B, or programmed death-ligand 1 (PD-L1). In summary, despite ERK inhibition by both U0126 and trametinib, only U0126 protected against AKI suggesting that the protection against AKI by U0126 was due to an off-target effect independent of ERK inhibition. The effect of U0126 to decrease AKI may be mediated by inhibition of heat shock protein 1, CDK4 or stratifin (14-3-3σ). Trametinib was more effective than cisplatin in decreasing tumor growth, but unlike cisplatin, trametinib did not cause AKI.
Collapse
Affiliation(s)
- Carolyn N Brown
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA
| | - Daniel J Atwood
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA
| | - Deepak Pokhrel
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA
| | - Kameswaran Ravichandran
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Sara J Holditch
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA
| | - Sanskriti Saxena
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA
| | - Makoto Miyazaki
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA
| | - Raphael Nemenoff
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA
| | - Mary C M Weiser-Evans
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA
| | | | - Melanie S Joy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Univ. of Colorado at Denver, Aurora, CO, USA
| | - Charles L Edelstein
- Division of Renal Diseases and Hypertension, Univ. of Colorado at Denver, Aurora, CO, USA.
| |
Collapse
|
28
|
Fontecha-Barriuso M, Martin-Sanchez D, Martinez-Moreno JM, Monsalve M, Ramos AM, Sanchez-Niño MD, Ruiz-Ortega M, Ortiz A, Sanz AB. The Role of PGC-1α and Mitochondrial Biogenesis in Kidney Diseases. Biomolecules 2020; 10:biom10020347. [PMID: 32102312 PMCID: PMC7072614 DOI: 10.3390/biom10020347] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the fastest growing causes of death worldwide, emphasizing the need to develop novel therapeutic approaches. CKD predisposes to acute kidney injury (AKI) and AKI favors CKD progression. Mitochondrial derangements are common features of both AKI and CKD and mitochondria-targeting therapies are under study as nephroprotective agents. PGC-1α is a master regulator of mitochondrial biogenesis and an attractive therapeutic target. Low PGC-1α levels and decreased transcription of its gene targets have been observed in both preclinical AKI (nephrotoxic, endotoxemia, and ischemia-reperfusion) and in experimental and human CKD, most notably diabetic nephropathy. In mice, PGC-1α deficiency was associated with subclinical CKD and predisposition to AKI while PGC-1α overexpression in tubular cells protected from AKI of diverse causes. Several therapeutic strategies may increase kidney PGC-1α activity and have been successfully tested in animal models. These include AMP-activated protein kinase (AMPK) activators, phosphodiesterase (PDE) inhibitors, and anti-TWEAK antibodies. In conclusion, low PGC-1α activity appears to be a common feature of AKI and CKD and recent characterization of nephroprotective approaches that increase PGC-1α activity may pave the way for nephroprotective strategies potentially effective in both AKI and CKD.
Collapse
Affiliation(s)
- Miguel Fontecha-Barriuso
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
| | - Diego Martin-Sanchez
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
| | - Julio Manuel Martinez-Moreno
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
| | - Maria Monsalve
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain;
| | - Adrian Mario Ramos
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
| | - Marta Ruiz-Ortega
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
- School of Medicine, UAM, 28029 Madrid, Spain
| | - Alberto Ortiz
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
- School of Medicine, UAM, 28029 Madrid, Spain
- IRSIN, 28040 Madrid, Spain
| | - Ana Belen Sanz
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-91-550-48-00
| |
Collapse
|
29
|
The effects of Hemiscorpius lepturus induced-acute kidney injury on PGC-1α gene expression: From induction to suppression in mice. Toxicon 2019; 174:57-63. [PMID: 31887316 DOI: 10.1016/j.toxicon.2019.12.154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/05/2019] [Accepted: 12/21/2019] [Indexed: 11/23/2022]
Abstract
Hemiscorpius lepturus envenomation induces acute kidney injury (AKI) through hemoglubinoria and mitochondrial dysfunction. Mitochondria supports ATP production to promote the regulation of fluid and electrolyte balance. Mitochondrial homeostasis in different metabolic environments can be adjusted by overexpression of PGC-1α. High reactive oxygen species (ROS) production after H. lepturus envenomation and heme oxygenase-1 (HO-1) overexpression causes ATP depletion as well as mitochondrial homeostasis disruption, which lead to progression in renal diseases. The present study aims to evaluate the role of venom induced-AKI in modulating mitochondrial function in cell death and metabolic signaling associated with PPAR-α, PGC-1α, and Nrf-2 as the main transcription factors involved in metabolism. Based on the data, two significant events occurred after envenomation: reduction of gl glutathione level and overexpression of the cytoprotective enzyme HO-1. Apaoptosis induction is associated with a significant decrease in the transcription of PPAR-α, PGC-1α and Nrf-2 after administrating lethal dose of venom (10 mg/kg). Furthermore, at the lower doses of venom (1 and 5 mg/kg), with a significant recovery accompanied with PGC-1α upregulation occurs after AKI. As the findings indicate, PGC-1α has a key role in restoring the mitochondrial function at the recovery phase of mouse model of AKI, which highlights the PGC-1α as a therapeutic target for venom induced-AKI prevention and treatment.
Collapse
|
30
|
Unexpected mechanism of colitis amelioration by artesunate, a natural product from Artemisia annua L. Inflammopharmacology 2019; 28:851-868. [PMID: 31865495 DOI: 10.1007/s10787-019-00678-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/28/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Artemisinin and its derivatives are known to exert immunosuppressive effects through modulating adaptive immunity. We investigated a novel role of artesunate in regulating innate immunity, including both macrophages (MΦ) and dendritic cells (DCs), which are known to involve in DSS-induced colitis. METHODS Effects of artesunate on innate immunity were extensively evaluated, both in vivo using DSS-colitis model with WT and T cell-deficient RAG mice (RAG-/-) and in vitro using cell culture models, including in-depth analyses of MΦ/DC apoptosis and cytokine expression by flow cytometry, Western blot, or immunohistology. RESULTS Unexpectedly, artesunate significantly ameliorated the DSS colitis of both WT and RAG1-/- mice with similar potency, suggesting a mechanism that involves primarily innate rather than adaptive immunity. In vivo mechanistic studies revealed that artesunate markedly induced apoptosis of lamina propria MΦs and DCs and suppressed mucosal TNF-α and IL-12p70 in DSS-colitis. In vitro, artesunate potently induced a dose- and time-dependent apoptosis of murine bone marrow-derived DCs and human THP-1 MΦs, through the caspases-9-mediated intrinsic pathway. Artesunate significantly decreased the secretion of IL-12p40/70 by DCs and TNF-α by MΦs. Furthermore, a combination of artesunate with an immunomodulator (methotrexate/triptolide/azathioprine) exhibited superior potency in promoting apoptosis of MΦs than any individual drug alone. CONCLUSIONS The immunomodulatory mechanism of artesunate in colitis involves a novel and potent induction of the intrinsic apoptosis pathway of proliferating MΦs and DCs and suppression of IL-12 and TNF-α. Artemisinin and its derivatives are promising new therapeutic alternatives for IBD, either alone or in combination with other immunomodulators.
Collapse
|
31
|
Huang W, Liu C, Xie L, Wang Y, Xu Y, Li Y. Integrated network pharmacology and targeted metabolomics to reveal the mechanism of nephrotoxicity of triptolide. Toxicol Res (Camb) 2019; 8:850-861. [PMID: 32110379 PMCID: PMC7017871 DOI: 10.1039/c9tx00067d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/06/2019] [Indexed: 12/21/2022] Open
Abstract
Triptolide (TP) is one of the important active components in Tripterygium wilfordii Hook. F., which shows strong anti-inflammatory and immunomodulatory effects. However, a large number of literature studies have reported that TP is the main component causing nephrotoxicity, and the mechanism of nephrotoxicity has not yet been revealed. Therefore, it is of great practical significance to clarify the toxicity mechanism of TP. This study integrated network pharmacology and targeted metabolomics to reveal the nephrotoxicity mechanism of TP. Firstly, network pharmacology screening of 61 action targets related to TP induced nephrotoxicity, with 39 direct targets and 22 indirect targets, was performed. Subsequently, based on a large-scale protein-protein interaction (PPI) and molecular docking validation, the core targets were identified. Based on the above targets and enrichment analysis, the purine metabolism, Toll-like receptor signaling pathway and NF-κB signaling pathway were found play a pivotal role in TP-induced nephrotoxicity. Literature investigation showed that purine and pyrimidine metabolism pathways were closely related to kidney diseases. Therefore, by using the quantitative method of determining endogenous purine and pyrimidine previously established in the laboratory, a targeted metabolomic analysis of TP was carried out. Finally, six nephrotoxicity biomarkers, dihydroorotate, thymidine, 2-deoxyinosine, uric acid, adenosine and xanthine, were found. Combining the above results, the mechanisms underlying the nephrotoxicity of TP were speculated to be due to the over-consumption of xanthine and uric acid, which would result in enormous ROS being released in response to oxidative stress in the body. Furthermore, activation of the Toll-like receptor signalling pathway can promotes the phosphorylation of the downstream protein NF-κB and causes an inflammatory response that ultimately leads to nephrotoxicity.
Collapse
Affiliation(s)
- Wei Huang
- School of Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , Jian Kang Chan Ye Yuan , Jinghai Dist. , Tianjin 301617 , China . ; ; ; Tel: +86-22-59596223
| | - Chuanxin Liu
- School of Chinese Materia Medica , Beijing University of Chinese Medicine , Liangxiang Town , Fangshan District , Beijing 102488 , China
| | - Lijuan Xie
- School of Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , Jian Kang Chan Ye Yuan , Jinghai Dist. , Tianjin 301617 , China . ; ; ; Tel: +86-22-59596223
| | - Yuming Wang
- School of Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , Jian Kang Chan Ye Yuan , Jinghai Dist. , Tianjin 301617 , China . ; ; ; Tel: +86-22-59596223
| | - Yanyan Xu
- School of Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , Jian Kang Chan Ye Yuan , Jinghai Dist. , Tianjin 301617 , China . ; ; ; Tel: +86-22-59596223
| | - Yubo Li
- School of Chinese Materia Medica , Tianjin University of Traditional Chinese Medicine , Jian Kang Chan Ye Yuan , Jinghai Dist. , Tianjin 301617 , China . ; ; ; Tel: +86-22-59596223
| |
Collapse
|
32
|
Lee G, Uddin MJ, Kim Y, Ko M, Yu I, Ha H. PGC-1α, a potential therapeutic target against kidney aging. Aging Cell 2019; 18:e12994. [PMID: 31313501 PMCID: PMC6718532 DOI: 10.1111/acel.12994] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is defined as changes in an organism over time. The proportion of the aged population is markedly increasing worldwide. The kidney, as an essential organ with a high energy requirement, is one of the most susceptible organs to aging. It is involved in glucose metabolism via gluconeogenesis, glucose filtration and reabsorption, and glucose utilization. Proximal tubular epithelial cells (PTECs) depend on lipid metabolism to meet the high demand for ATP. Recent studies have shown that aging‐related kidney dysfunction is highly associated with metabolic changes in the kidney. Peroxisome proliferator‐activated receptor gamma coactivator‐1 alpha (PGC‐1α), a transcriptional coactivator, plays a major role in the regulation of mitochondrial biogenesis, peroxisomal biogenesis, and glucose and lipid metabolism. PGC‐1α is abundant in tissues, including kidney PTECs, which demand high energy. Many in vitro and in vivo studies have demonstrated that the activation of PGC‐1α by genetic or pharmacological intervention prevents telomere shortening and aging‐related changes in the skeletal muscle, heart, and brain. The activation of PGC‐1α can also prevent kidney dysfunction in various kidney diseases. Therefore, a better understanding of the effect of PGC‐1α activation in various organs on aging and kidney diseases may unveil a potential therapeutic strategy against kidney aging.
Collapse
Affiliation(s)
- Gayoung Lee
- Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Md Jamal Uddin
- Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Yoojeong Kim
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Minji Ko
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Inyoung Yu
- College of Pharmacy Ewha Womans University Seoul Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences Ewha Womans University Seoul Korea
- College of Pharmacy Ewha Womans University Seoul Korea
| |
Collapse
|
33
|
Heme Oxygenase-1 Protects the Liver from Septic Injury by Modulating TLR4-Mediated Mitochondrial Quality Control in Mice. Shock 2019; 50:209-218. [PMID: 29028772 DOI: 10.1097/shk.0000000000001020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mitochondrial dysfunction is involved in the pathogenesis of sepsis-induced multiple organ dysfunction syndrome (MODS). Mitochondrial quality control (QC) is characterized by self-recovering mitochondrial damage through mitochondrial biogenesis, mitophagy, and fission/fusion. Heme oxygenase (HO)-1 acts as a signaling molecule to modulate inflammation. The present study elucidated the cytoprotective mechanisms of HO-1 in sepsis, particularly focusing on toll-like receptor (TLR)4-mediated mitochondrial QC. Mice were subjected to sepsis by cecal ligation and puncture (CLP). The mice were injected intraperitoneally with hemin (10 mg/kg) at 12 h before CLP or zinc protoporphyrin IX (ZnPP; 30 mg/kg) at 2 h before CLP. The serum and tissues were collected 6 h after CLP. Mortality, MODS, and proinflammatory cytokines increased in septic mice. These increases were augmented by ZnPP but attenuated by hemin. Hemin decreased mitochondrial lipid peroxidation and mitochondrial dysfunction. Hemin enhanced mitochondrial biogenesis, as indicated by increased levels of peroxisome proliferator-activated receptor-γ coactivator 1α, nuclear respiratory factor 1, and mitochondrial transcription factor A (TFAM). Hemin also enhanced mitophagy, as indicated by decreased PTEN-induced putative kinase 1 (PINK1) level and increased Parkin level. Hemin decreased fission-related protein, dynamin-related protein 1 (DRP1), and increased fusion-related protein, mitofusin 2. Hemin attenuated the increased TLR4 expression. TAK-242, a TLR4 antagonist, attenuated mortality, inflammatory response, and impaired mitochondrial QC. Our findings suggest that HO-1 attenuates septic injury by modulating TLR4-mediated mitochondrial QC.
Collapse
|
34
|
E6 Oncoproteins from High-Risk Human Papillomavirus Induce Mitochondrial Metabolism in a Head and Neck Squamous Cell Carcinoma Model. Biomolecules 2019; 9:biom9080351. [PMID: 31398842 PMCID: PMC6722992 DOI: 10.3390/biom9080351] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) cells that are positive for human papillomavirus (HPV+) favor mitochondrial metabolism rather than glucose metabolism. However, the involvement of mitochondrial metabolism in HNSCC HPV+ cells is still unknown. The aim of this work was to evaluate the role of E6 oncoproteins from HPV16 and HPV18 in the mitochondrial metabolism in an HNSCC model. We found that E6 from both viral types abates the phosphorylation of protein kinase B-serine 473 (pAkt), which is associated with a shift in mitochondrial metabolism. E6 oncoproteins increased the levels of protein subunits of mitochondrial complexes (I to IV), as well as the ATP synthase and the protein levels of the voltage dependent anion channel (VDAC). Although E6 proteins increased the basal and leak respiration, the ATP-linked respiration was not affected, which resulted in mitochondrial decoupling. This increase in leak respiration was associated to the induction of oxidative stress (OS) in cells expressing E6, as it was observed by the fall in the glutathione/glutathione disulfide (GSH/GSSG) rate and the increase in reactive oxygen species (ROS), carbonylated proteins, and DNA damage. Taken together, our results suggest that E6 oncoproteins from HPV16 and HPV18 are inducers of mitochondrial metabolism.
Collapse
|
35
|
Lee H, Oh S, Yang W, Park R, Kim H, Jeon JS, Noh H, Han DC, Cho KW, Kim YJ, Kwon SH. Bariatric Surgery Reduces Elevated Urinary Mitochondrial DNA Copy Number in Patients With Obesity. J Clin Endocrinol Metab 2019; 104:2257-2266. [PMID: 30657970 DOI: 10.1210/jc.2018-01935] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/11/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Obesity is an independent risk factor for chronic kidney disease. Recently, urinary mitochondrial DNA (mtDNA) has been used as a surrogate marker of mitochondrial damage in various kidney diseases. However, there are no data regarding its use in patients with obesity or the change in urinary mtDNA copy number after surgery. DESIGN We prospectively recruited age- and sex-matched healthy volunteers and patients with obesity (n = 22 in each group: nine men and 13 women). The copy number of urinary and serum mtDNA nicotinamide adenine dinucleotide dehydrogenase subunit-1 (mtND-1) and cytochrome-c oxidase 3 (mtCOX-3) was measured using quantitative PCR. We measured urinary mtDNA and body weight and carried out laboratory tests, 6 months after surgery. RESULTS Urinary mtND-1 copy number was significantly higher in the obese group than in healthy volunteers. However, urinary mtCOX-3 and serum ND-1 copy numbers in the obese group did not differ from that in the healthy volunteers. When patients with obesity were divided into two groups, according to their baseline mtND-1 copy number, bariatric surgery reduced the mtND-1 copy number (P = 0.006) in the high baseline mtDNA copy-number group. The change in urinary mtND-1 copy number was correlated with a change in urinary albumin (r = 0.478, P = 0.025). CONCLUSIONS Obesity is associated with elevated urinary mtND-1 copy number. Bariatric surgery reduces the elevated urinary mtND-1 copy number in patients with obesity. This suggests that bariatric surgery could attenuate mitochondrial damage in the kidney cells of patients with obesity.
Collapse
Affiliation(s)
- Haekyung Lee
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Songhee Oh
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Wonmi Yang
- Hyonam Kidney Laboratory, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Rojin Park
- Department of Laboratory Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hyoungnae Kim
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Jin Seok Jeon
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
- Hyonam Kidney Laboratory, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hyunjin Noh
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
- Hyonam Kidney Laboratory, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Dong Cheol Han
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
- Hyonam Kidney Laboratory, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Kae Won Cho
- Soonchunhyang Institute of Med-Bio Science, Soonchunhyang University, Cheonan, Chungchung nam do, Republic of Korea
| | - Yong Jin Kim
- Department of Surgery, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Soon Hyo Kwon
- Division of Nephrology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
- Hyonam Kidney Laboratory, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| |
Collapse
|
36
|
The Protective Effect of Alpha-Mangostin against Cisplatin-Induced Cell Death in LLC-PK1 Cells is Associated to Mitochondrial Function Preservation. Antioxidants (Basel) 2019; 8:antiox8050133. [PMID: 31096625 PMCID: PMC6562511 DOI: 10.3390/antiox8050133] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/28/2022] Open
Abstract
Cis-dichlorodiammineplatinum II (CDDP) is a chemotherapeutic agent that induces nephrotoxicity by different mechanisms, including oxidative stress, mitochondrial dysfunction, autophagy, and endoplasmic reticulum stress. This study aimed to evaluate if the protective effects of the antioxidant alpha-mangostin (αM) in CDDP-induced damage in proximal tubule Lilly laboratory culture porcine kidney (LLC-PK1) cells, are related to mitochondrial function preservation. It was found that αM co-incubation prevented CDDP-induced cell death. Furthermore, αM prevented the CDDP-induced decrease in cell respiratory states, in the maximum capacity of the electron transfer system (E) and in the respiration associated to oxidative phosphorylation (OXPHOS). CDDP also decreased the protein levels of voltage dependence anion channel (VDAC) and mitochondrial complex subunits, which together with the reduction in E, the mitofusin 2 decrease and the mitochondrial network fragmentation observed by MitoTracker Green, suggest the mitochondrial morphology alteration and the decrease in mitochondrial mass induced by CDDP. CDDP also induced the reduction in mitochondrial biogenesis observed by transcription factor A, mitochondria (TFAM) decreased protein-level and the increase in mitophagy. All these changes were prevented by αM. Taken together, our results imply that αM’s protective effects in CDDP-induced toxicity in LLC-PK1 cells are associated to mitochondrial function preservation.
Collapse
|
37
|
Wu Y, Yao YM, Lu ZQ. Mitochondrial quality control mechanisms as potential therapeutic targets in sepsis-induced multiple organ failure. J Mol Med (Berl) 2019; 97:451-462. [PMID: 30788535 DOI: 10.1007/s00109-019-01756-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 12/24/2018] [Accepted: 02/06/2019] [Indexed: 02/07/2023]
Abstract
Sepsis is a dysregulated response to severe infection characterized by life-threatening organ failure and is the leading cause of mortality worldwide. Multiple organ failure is the central characteristic of sepsis and is associated with poor outcome of septic patients. Ultrastructural damage to the mitochondria and mitochondrial dysfunction are reported in sepsis. Mitochondrial dysfunction with subsequent ATP deficiency, excessive reactive oxygen species (ROS) release, and cytochrome c release are all considered to contribute to organ failure. Consistent mitochondrial dysfunction leads to reduced mitochondrial quality control capacity, which eliminates dysfunctional and superfluous mitochondria to maintain mitochondrial homeostasis. Mitochondrial quality is controlled through a series of processes including mitochondrial biogenesis, mitochondrial dynamics, mitophagy, and transport processes. Several studies have indicated that multiple organ failure is ameliorated by restoring mitochondrial quality control mechanisms and is further amplified by defective quality control mechanisms. This review will focus on advances concerning potential mechanisms in regulating mitochondrial quality control and impacts of mitochondrial quality control on the progression of sepsis.
Collapse
Affiliation(s)
- You Wu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China.,Wenzhou Municipal Key Laboratory of Emergency, Critical Care and Disaster Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yong-Ming Yao
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China. .,Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China.
| | - Zhong-Qiu Lu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China. .,Wenzhou Municipal Key Laboratory of Emergency, Critical Care and Disaster Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China. .,College of Nursing, Wenzhou Medical University, Wenzhou, People's Republic of China.
| |
Collapse
|
38
|
Increased TLR4 Expression Aggravates Sepsis by Promoting IFN- γ Expression in CD38 -/- Mice. J Immunol Res 2019; 2019:3737890. [PMID: 30915370 PMCID: PMC6399547 DOI: 10.1155/2019/3737890] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/07/2018] [Accepted: 12/02/2018] [Indexed: 12/23/2022] Open
Abstract
Gram-negative bacterial sepsis accounts for up to 50% worldwide sepsis that causes hospital mortality. Acute kidney injury (AKI), a common complication of Gram-negative bacterial sepsis, is caused by Toll-like receptor 4 (TLR4) activation. Lipopolysaccharide (LPS) is an endotoxin in Gram-negative bacteria and is recognized specifically by TLR4, which initiates innate immune response. Also, TLR4 signaling pathway activation is essential in response to LPS infection. CD38 is one of the well-known regulators of innate immunity, whose dysregulation contributes to sepsis. Many studies have proven that an attenuated Gram-positive bacterium induces sepsis in a CD38-blocking model. However, the pathogenesis of Gram-negative bacteria-induced sepsis in a CD38−/− mouse model remains unclear. The aim of this study is to investigate whether kidney injury is still attenuated in a LPS-induced CD38−/− sepsis model and identify the potential mechanism. We assess the severity of kidney injury related to proinflammatory cytokine expressions (IFN-γ, TNF-α, IL-1β, and IL-6) in WT and CD38−/− mice. Our results showed more aggravated kidney damage in CD38−/− mice than in WT mice, accompanied with an increase of proinflammatory cytokine expression. In addition, compared with CD38−/−TLR4mut mice, we found an increase of TLR4 expression and mRNA expression of these cytokines in the kidney of CD38−/− mice, although only increased IFN-γ level was detected in the serum. Taken together, these results demonstrated that an increased TLR4 expression in CD38−/− mice could contribute to the aggravation of AKI through boosting of the production of IFN-γ.
Collapse
|
39
|
Zynda ER, Maloy MH, Kandel ES. The role of PAK1 in the sensitivity of kidney epithelial cells to ischemia-like conditions. Cell Cycle 2019; 18:596-604. [PMID: 30724698 DOI: 10.1080/15384101.2019.1578149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Kidney ischemia, characterized by insufficient supply of oxygen and nutrients to renal epithelial cells, is the main cause of acute kidney injury and an important contributor to mortality world-wide. Earlier research implicated a G-protein coupled receptor (NK1R) in the death of kidney epithelial cells in ischemia-like conditions. P21-associated kinase 1 (PAK1) is involved in signalling by several G-proteins. We explored the consequences of PAK1 inhibition for cell survival under the conditions of reduced glucose and oxygen. Inhibition of PAK1 by RNA interference, expression of a dominant-negative mutant or treatment with small molecule inhibitors greatly reduced the death of cultured kidney epithelial cells. Similar protection was achieved by treating the cells with inhibitors of MEK1, in agreement with the prior reports on PAK1-MEK1 connection. Concomitant inhibition of NK1R and PAK1 offered no better protection than inhibition of NK1R alone, consistent with the two proteins being members of the same pathway. Furthermore, NK1R, PAK and MEK inhibitors reduced the induction of TRAIL in ischemia-like conditions. Considering the emerging role of TRAIL in ischemia-mediated cell death, this phenomenon may contribute to the protective effects of these small molecules. Our findings support further exploration of PAK and MEK inhibitors as possible agents to avert ischemic kidney injury.
Collapse
Affiliation(s)
- Evan R Zynda
- a Department of Cell Stress Biology , Roswell Park Comprehensive Cancer Center , Buffalo , NY , USA
| | - Mitchell H Maloy
- a Department of Cell Stress Biology , Roswell Park Comprehensive Cancer Center , Buffalo , NY , USA
| | - Eugene S Kandel
- a Department of Cell Stress Biology , Roswell Park Comprehensive Cancer Center , Buffalo , NY , USA
| |
Collapse
|
40
|
Netti GS, Sangregorio F, Spadaccino F, Staffieri F, Crovace A, Infante B, Maiorano A, Godeas G, Castellano G, Di Palma AM, Prattichizzo C, Cotoia A, Mirabella L, Gesualdo L, Cinnella G, Stallone G, Ranieri E, Grandaliano G. LPS removal reduces CD80-mediated albuminuria in critically ill patients with Gram-negative sepsis. Am J Physiol Renal Physiol 2019; 316:F723-F731. [PMID: 30672713 DOI: 10.1152/ajprenal.00491.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
LPS-induced sepsis is a leading cause of acute kidney injury (AKI) in critically ill patients. LPS may induce CD80 expression in podocytes with subsequent onset of proteinuria, a risk factor for progressive chronic kidney disease (CKD) frequently observed after AKI. This study aimed to investigate the therapeutic efficacy of LPS removal in decreasing albuminuria through the reduction of podocyte CD80 expression. Between January 2015 and December 2017, 70 consecutive patients with Gram-negative sepsis-induced AKI were randomized to either have coupled plasma filtration and adsorption (CPFA) added to the standard care ( n = 35) or not ( n = 35). To elucidate the possible relationship between LPS-induced renal damage, proteinuria, and CD80 expression in Gram sepsis, a swine model of LPS-induced AKI was set up. Three hours after LPS infusion, animals were treated or not with CPFA for 6 h. Treatment with CPFA significantly reduced serum cytokines, C-reactive protein, procalcitonin, and endotoxin levels in patients with Gram-negative sepsis-induced AKI. CPFA significantly lowered also proteinuria and CD80 urinary excretion. In the swine model of LPS-induced AKI, CD80 glomerular expression, which was undetectable in control pigs, was markedly increased at the podocyte level in LPS-exposed animals. CPFA significantly reduced LPS-induced proteinuria and podocyte CD80 expression in septic pigs. Our data indicate that LPS induces albuminuria via podocyte expression of CD80 and suggest a possible role of timely LPS removal in preventing the maladaptive repair of the podocytes and the consequent increased risk of CKD in sepsis-induced AKI.
Collapse
Affiliation(s)
- Giuseppe Stefano Netti
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Fabio Sangregorio
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Federica Spadaccino
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Francesco Staffieri
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro , Bari , Italy
| | - Antonio Crovace
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro , Bari , Italy
| | - Barbara Infante
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Annamaria Maiorano
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Giulia Godeas
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Giuseppe Castellano
- Nephrology Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro , Bari , Italy
| | - Anna Maria Di Palma
- Nephrology Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro , Bari , Italy
| | - Clelia Prattichizzo
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Antonella Cotoia
- Anesthesia and Intensive Care Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Lucia Mirabella
- Anesthesia and Intensive Care Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Loreto Gesualdo
- Nephrology Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro , Bari , Italy
| | - Gilda Cinnella
- Anesthesia and Intensive Care Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Giovanni Stallone
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Elena Ranieri
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| | - Giuseppe Grandaliano
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia , Foggia , Italy
| |
Collapse
|
41
|
Dik B, Sonmez G, Faki HE, Bahcivan E. Sulfasalazine treatment can cause a positive effect on LPS-induced endotoxic rats. Exp Anim 2018; 67:403-412. [PMID: 29731490 PMCID: PMC6219878 DOI: 10.1538/expanim.18-0029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The aim of this study, was to determine the effect of sulfasalazine for different periods
of time reduces disseminated intravascular coagulation, inflammation and organ damages by
inhibiting the nuclear factor kappa beta pathway. The study was performed with 30 Wistar
albino rats and the groups were established as Control group, LPS group; endotoxemia was
induced with LPS, SL5 group: sulfasalazine (300 mg/kg, single dose daily) was administered
for 5 days before the LPS-induced endotoxemia, and LS group: sulfasalazine (300 mg/kg,
single dose) was administered similtenously with LPS. Hemogram, biochemical, cytokine
(IL-1β, IL-6, IL-10, TNF-α) and acute phase proteins (HPT, SAA, PGE2) analyzes and
oxidative status values were measured from blood samples at 3 and 6 h after the last
applications in the all groups. The rats were euthanized at 6 h and mRNA
levels of BCL2 and BAX genes were examined from liver
and brain tissues. Sulfasalazine reduced the increased IL-1β, IL-6, TNF-α and
PGE2 levels and significantly increased anti-inflammatory cytokine IL-10
levels. In addition, decreasing of ATIII level was prevented in the SL5 group, and
decreasing of fibrinogen levels were prevented in the LS and SL5 groups within first 3 h.
In LPS group, leukocyte and thrombocyte levels were decreased, however sulfasalazine
application inhibited decreases of leukocyte levels in LS and SL5 groups. In addition,
sulfasalazine inhibited the decrease of total antioxidant capacity and unchanged apoptosis
in brain and liver. In conclusion, the use of sulfasalazine in different durations reduce
the excessive inflammation of endotoxemia cases.
Collapse
Affiliation(s)
- Burak Dik
- Department of Pharmacology and Toxicology, Veterinary Faculty, Selcuk University, New Istanbul Highway, 42130 Konya, Turkey
| | - Gonca Sonmez
- Department of Genetics, Veterinary Faculty, Selcuk University, New Istanbul Highway, 42130 Konya, Turkey
| | - Hatice Eser Faki
- Department of Pharmacology and Toxicology, Veterinary Faculty, Selcuk University, New Istanbul Highway, 42130 Konya, Turkey
| | - Emre Bahcivan
- Department of Pharmacology and Toxicology, Veterinary Faculty, Kafkas University, 36300 Kars, Turkey
| |
Collapse
|
42
|
Chuang CL, Chang CC, Hsu SJ, Huang HC, Lee FY, Huang LJ, Lee SD. Endotoxemia-enhanced renal vascular reactivity to endothelin-1 in cirrhotic rats. Am J Physiol Gastrointest Liver Physiol 2018; 315:G752-G761. [PMID: 30095297 DOI: 10.1152/ajpgi.00302.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatorenal syndrome (HRS), a severe complication of advanced cirrhosis, is defined as hypoperfusion of kidneys resulting from intense renal vasoconstriction in response to generalized systemic arterial vasodilatation. Nevertheless, the mechanisms have been barely investigated. Cumulative studies demonstrated renal vasodilatation in portal hypertensive and compensated cirrhotic rats. Previously, we identified that blunted renal vascular reactivity of portal hypertensive rats was reversed after lipopolysaccharide (LPS). This study was therefore conducted to delineate the sequence of renal vascular alternation and underlying mechanisms in LPS-treated cirrhotic rats. Sprague-Dawley rats were randomly allocated to receive sham surgery (Sham) or common bile duct ligation (CBDL). LPS was induced on the 28th day after surgery. Kidney perfusion was performed at 0.5 or 3 h after LPS to evaluate renal vascular response to endothelin-1 (ET-1). Endotoxemia increased serum ET-1 levels ( P < 0.0001) and renal arterial blood flow ( P < 0.05) in both Sham and CBDL rats. CBDL rats showed enhanced renal vascular reactivity to ET-1 at 3 h after LPS ( P = 0.026). Pretreatment with endothelin receptor type A (ETA) antagonist abrogated the LPS-enhanced renal vascular response in CBDL rats ( P < 0.001). There were significantly lower inducible nitric oxide synthase (iNOS) expression but higher ETA and phosphorylated extracellular signal-regulated kinase (p-ERK) expressions in renal medulla of endotoxemic CBDL rats ( P < 0.05). We concluded that LPS-induced renal iNOS inhibition, ETA upregulation, and subsequent ERK signaling activation may participate in renal vascular hyperreactivity in cirrhosis. ET-1-targeted therapy may be feasible in the control of HRS. NEW & NOTEWORTHY Hepatorenal syndrome (HRS) occurred in advanced cirrhosis after large-volume paracentesis or bacterial peritonitis. We demonstrated that intraperitoneal lipopolysaccharide (LPS) enhanced renal vascular reactivity to endothelin-1 (ET-1) in cirrhotic rats, accompanied by inducible nitric oxide synthase inhibition, endothelin receptor type A (ETA) upregulation, and subsequent extracellular signal-regulated kinase activation in renal medulla. Pretreatment with ETA antagonist abrogated the LPS-enhanced renal vascular response in common bile duct ligation rats. These findings suggest that further clinical investigation of ET-1-targeted therapy may be feasible in the control of HRS.
Collapse
Affiliation(s)
- Chiao-Lin Chuang
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital , Taipei , Taiwan.,Faculty of Medicine, National Yang-Ming University School of Medicine , Taipei , Taiwan
| | - Ching-Chih Chang
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital , Taipei , Taiwan.,Faculty of Medicine, National Yang-Ming University School of Medicine , Taipei , Taiwan
| | - Shao-Jung Hsu
- Faculty of Medicine, National Yang-Ming University School of Medicine , Taipei , Taiwan.,Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital , Taipei , Taiwan
| | - Hui-Chun Huang
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital , Taipei , Taiwan.,Faculty of Medicine, National Yang-Ming University School of Medicine , Taipei , Taiwan.,Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital , Taipei , Taiwan
| | - Fa-Yauh Lee
- Faculty of Medicine, National Yang-Ming University School of Medicine , Taipei , Taiwan.,Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital , Taipei , Taiwan
| | - Ling-Ju Huang
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital , Taipei , Taiwan.,Faculty of Medicine, National Yang-Ming University School of Medicine , Taipei , Taiwan.,Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital , Taipei , Taiwan
| | - Shou-Dong Lee
- Faculty of Medicine, National Yang-Ming University School of Medicine , Taipei , Taiwan.,Division of Gastroenterology, Department of Medicine, Cheng-Hsin General Hospital , Taipei , Taiwan
| |
Collapse
|
43
|
Yan HP, Li M, Lu XL, Zhu YM, Ou-Yang WX, Xiao ZH, Qiu J, Li SJ. Use of plasma mitochondrial DNA levels for determining disease severity and prognosis in pediatric sepsis: a case control study. BMC Pediatr 2018; 18:267. [PMID: 30092777 PMCID: PMC6085664 DOI: 10.1186/s12887-018-1239-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 07/31/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The mortality rate due to severe sepsis is approximately 30-60%. Sepsis readily progresses to septic shock and multiple organ dysfunction, representing a significant problem in the pediatric intensive care unit (PICU). The aim of this study was to explore the value of plasma mitochondrial DNA (mtDNA) for early diagnosis and prognosis in children with sepsis. METHODS A total of 123 children with sepsis who were hospitalized in the Hunan Children's Hospital PICU from July 2013 to December 2014 were divided into the general sepsis group (n = 70) and severe sepsis group (n = 53) based on diagnostic standards. An additional 30 children with non-sepsis infection and 30 healthy children were randomly selected as a control group. Patients' plasma was collected during admission to the PICU. A pediatric critical illness score (PCIS) was also calculated. The plasma mtDNA level was examined using real-time polymerase chain reaction technology, and other parameters including routine laboratory values; blood lactate, procalcitonin (PCT), and C-reactive protein (CRP) levels; and data on survival were collected and compared among the groups. RESULTS The plasma mtDNA level in the sepsis group than that in the non-sepsis infection and healthy groups. The plasma mtDNA level was significantly higher in the severe sepsis than in the general sepsis group (p < 0.001). A lower PCIS was associated with a higher plasma mtDNA level (p < 0.001). A higher number of organs with dysfunction was associated with higher plasma mtDNA levels (p < 0.001). Plasma mtDNA levels were higher among patients with elevated alanine aminotransferase, aspartate aminotransferase, blood urea nitrogen, creatinine, lactate dehydrogenase, creatine kinase, myoglobin, creatine kinase MB, and troponin than in those with values within the normal range. The mtDNA level was higher among non-survivors than among survivors, and this difference was significant. mtDNA showed a prognostic prediction value similar to that of lactate, PCT, and CRP. CONCLUSIONS Plasma mtDNA levels may be a suitable biomarker for diagnosis and prognosis in children with sepsis.
Collapse
Affiliation(s)
- Hai Peng Yan
- Department of Pediatric Intensive Care Unit (PICU), Hunan Children's Hospital, Changsha, China
| | - Miao Li
- Department of Pediatric Intensive Care Unit (PICU), Hunan Children's Hospital, Changsha, China
| | - Xiu Lan Lu
- Department of Pediatric Intensive Care Unit (PICU), Hunan Children's Hospital, Changsha, China
| | - Yi Min Zhu
- Hunan Provincial People's Hospital, the first affiliated hospital of Hunan normal University, Changsha, 410007, People's Republic of China
| | - Wen-Xian Ou-Yang
- Department of Section of Liver Disease, Hunan Children's Hospital, 86# Ziyuan Road, Changsha, 410007, China
| | - Zheng Hui Xiao
- Department of Pediatric Intensive Care Unit (PICU), Hunan Children's Hospital, Changsha, China
| | - Jun Qiu
- Department of Pediatric Intensive Care Unit (PICU), Hunan Children's Hospital, Changsha, China
| | - Shuang Jie Li
- Department of Section of Liver Disease, Hunan Children's Hospital, 86# Ziyuan Road, Changsha, 410007, China.
| |
Collapse
|
44
|
Bryant JL, Guda PR, Asemu G, Subedi R, Ray S, Khalid OS, Shukla V, Patel D, Davis H, Nimmagadda VKC, Makar TK. Glomerular mitochondrial changes in HIV associated renal injury. Exp Mol Pathol 2018; 104:175-189. [PMID: 29608912 DOI: 10.1016/j.yexmp.2018.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/29/2018] [Indexed: 12/20/2022]
Abstract
HIV-associated nephropathy (HIVAN) is an AIDs-related disease of the kidney. HIVAN is characterized by severe proteinuria, podocyte hyperplasia, collapse, glomerular, and tubulointerstitial damage. HIV-1 transgenic (Tg26) mouse is the most popular model to study the HIV manifestations that develop similar renal presentations as HIVAN. Viral proteins, including Tat, Nef, and Vpr play a significant role in renal cell damage. It has been shown that mitochondrial changes are involved in several kidney diseases, and therefore, mitochondrial dysfunction may be implicated in the pathology of HIVAN. In the present study, we investigated the changes of mitochondrial homeostasis, biogenesis, dynamics, mitophagy, and examined the role of reactive oxygen species (ROS) generation and apoptosis in the Tg26 mouse model. The Tg26 mice showed significant impairment of kidney function, which was accompanied by increased blood urea nitrogen (BUN), creatinine and protein urea level. In addition, histological, western blot and PCR analysis of the Tg26 mice kidneys showed a downregulation of NAMPT, SIRT1, and SIRT3 expressions levels. Furthermore, the kidney of the Tg26 mice showed a downregulation of PGC1α, MFN2, and PARKIN, which are coupled with decrease of mitochondrial biogenesis, imbalance of mitochondrial dynamics, and downregulation of mitophagy, respectively. Furthermore, our results indicate that mitochondrial dysfunction were associated with ER stress, ROS generation and apoptosis. These results strongly suggest that the impaired mitochondrial morphology, homeostasis, and function associated with HIVAN. These findings indicated that a new insight on pathological mechanism associated with mitochondrial changes in HIVAN and a potential therapeutic target.
Collapse
Affiliation(s)
- Joseph L Bryant
- Institute of Human Virology, University of Maryland, Baltimore, MD, United States
| | | | - Girma Asemu
- Institute of Human Virology, University of Maryland, Baltimore, MD, United States
| | - Rogin Subedi
- Department of Neurology, University of Maryland, Baltimore, MD, United States
| | - Sugata Ray
- Department of Neurology, University of Maryland, Baltimore, MD, United States
| | - Omar S Khalid
- Department of Neurology, University of Maryland, Baltimore, MD, United States
| | - Vivek Shukla
- Department of Neurology, University of Maryland, Baltimore, MD, United States
| | - Dhruvil Patel
- Department of Neurology, University of Maryland, Baltimore, MD, United States
| | - Harry Davis
- Institute of Human Virology, University of Maryland, Baltimore, MD, United States
| | | | - Tapas K Makar
- Department of Neurology, University of Maryland, Baltimore, MD, United States.
| |
Collapse
|
45
|
Van Beusecum JP, Zhang S, Cook AK, Inscho EW. Acute toll-like receptor 4 activation impairs rat renal microvascular autoregulatory behaviour. Acta Physiol (Oxf) 2017; 221:204-220. [PMID: 28544543 DOI: 10.1111/apha.12899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/16/2016] [Accepted: 05/18/2017] [Indexed: 12/24/2022]
Abstract
AIM Little is known about how toll-like receptor 4 (TLR4) influences the renal microvasculature. We hypothesized that acute TLR4 stimulation with lipopolysaccharide (LPS) impairs afferent arteriole autoregulatory behaviour, partially through reactive oxygen species (ROS). METHODS We assessed afferent arteriole autoregulatory behaviour after LPS treatment (1 mg kg-1 ; i.p.) using the in vitro blood-perfused juxtamedullary nephron preparation. Autoregulatory behaviour was assessed by measuring diameter responses to stepwise changes in renal perfusion pressure. TLR4 expression was assessed by immunofluorescence, immunohistochemistry and Western blot analysis in the renal cortex and vasculature. RESULTS Baseline arteriole diameter at 100 mmHg averaged 15.2 ± 1.2 μm and 12.2 ± 1.0 μm for control and LPS groups (P < 0.05) respectively. When perfusion pressure was increased in 15 mmHg increments from 65 to 170 mmHg, arteriole diameter in control kidneys decreased significantly to 69 ± 6% of baseline diameter. In the LPS-treated group, arteriole diameter remained essentially unchanged (103 ± 9% of baseline), indicating impaired autoregulatory behaviour. Pre-treatment with anti-TLR4 antibody or the TLR4 antagonist, LPS-RS, preserved autoregulatory behaviour during LPS treatment. P2 receptor reactivity was normal in control and LPS-treated rats. Pre-treatment with Losartan (angiotensin type 1 receptor blocker; (AT1 ) 2 mg kg-1 ; i.p.) increased baseline afferent arteriole diameter but did not preserve autoregulatory behaviour in LPS-treated rats. Acute exposure to Tempol (10-3 mol L-1 ), a superoxide dismutase mimetic, restored pressure-mediated vasoconstriction in kidneys from LPS-treated rats. CONCLUSION These data demonstrate that TLR4 activation impairs afferent arteriole autoregulatory behaviour, partially through ROS, but independently of P2 and AT1 receptor activation.
Collapse
Affiliation(s)
- J. P. Van Beusecum
- Division of Nephrology; Department of Medicine; University of Alabama at Birmingham; Birmingham AL USA
- Department of Physiology; Augusta University; Augusta GA USA
| | - S. Zhang
- Division of Nephrology; Department of Medicine; University of Alabama at Birmingham; Birmingham AL USA
- Department of Physiology; Augusta University; Augusta GA USA
| | - A. K. Cook
- Division of Nephrology; Department of Medicine; University of Alabama at Birmingham; Birmingham AL USA
- Department of Physiology; Augusta University; Augusta GA USA
| | - E. W. Inscho
- Division of Nephrology; Department of Medicine; University of Alabama at Birmingham; Birmingham AL USA
- Department of Physiology; Augusta University; Augusta GA USA
| |
Collapse
|
46
|
Collier JB, Schnellmann RG. Extracellular Signal-Regulated Kinase 1/2 Regulates Mouse Kidney Injury Molecule-1 Expression Physiologically and Following Ischemic and Septic Renal Injury. J Pharmacol Exp Ther 2017; 363:419-427. [PMID: 29074644 DOI: 10.1124/jpet.117.244152] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/27/2017] [Indexed: 01/31/2023] Open
Abstract
The upregulation of kidney injury molecule-1 (KIM-1) has been extensively studied in various renal diseases and following acute injury; however, the initial mechanisms controlling KIM-1 expression remain limited. In this study, KIM-1 expression was examined in mouse renal cell cultures and in two different models of acute kidney injury (AKI), ischemia reperfusion (IR)-induced and lipopolysaccharide (LPS)-induced sepsis. KIM-1 mRNA increased in both AKI models, and pharmacological inhibition of extracellular signal-regulated kinase 1/2 (ERK1/2) signaling attenuated injury-induced KIM-1 expression in the renal cortex. Toll-like receptor 4 knockout (TLR4KO) mice exhibited reduced ERK1/2 phosphorylation and attenuated KIM-1 mRNA after LPS exposure. TLR4KO mice were not protected from IR-induced ERK1/2 phosphorylation and upregulation of KIM-1 mRNA. Following renal IR injury, phosphorylation of signal transducer and activator of transcription 3 (STAT3) at serine 727 and tyrosine 705 increased downstream from ERK1/2 activation. Because phosphorylated STAT3 is a transcriptional upregulator of KIM-1 and inhibition of ERK1/2 attenuated increases in STAT3 phosphorylation, we suggest an ERK1/2-STAT3-KIM-1 pathway following renal injury. Finally, ERK1/2 inhibition in naive mice decreased KIM-1 mRNA and nuclear STAT3 phosphorylation in the cortex, indicating homeostatic regulation of KIM-1. These findings reveal renal ERK1/2 as an important initial regulator of KIM-1 expression in IR and septic AKI and at a physiologic level.Visual Abstract.Proposed mechanism of IR, LPS, and ROS-induced renal damage that initiates ERK1/2 and STAT3 phosphorylation. STAT3 then binds to the KIM-1 promoter and increases KIM-1 mRNA. By preventing ERK1/2 phosphorylation following renal injury, STAT3 phosphorylation is decreased, leading to less phosphorylated STAT3 within the nucleus, and subsequently less KIM-1 mRNA increases post injury.
Collapse
Affiliation(s)
- Justin B Collier
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (J.B.C.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (R.G.S.)
| | - Rick G Schnellmann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (J.B.C.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (R.G.S.)
| |
Collapse
|
47
|
Abstract
Acute kidney injury (AKI) arising from diverse etiologies is characterized by mitochondrial dysfunction. The peroxisome proliferator-activated receptor γ coactivator-1alpha (PGC1α), a master regulator of mitochondrial biogenesis, has been shown to be protective in AKI. Interestingly, reduction of PGC1α has also been implicated in the development of diabetic kidney disease and renal fibrosis. The beneficial renal effects of PGC1α make it a prime target for therapeutics aimed at ameliorating AKI, forms of chronic kidney disease (CKD), and their intersection. This review summarizes the current literature on the relationship between renal health and PGC1α and proposes areas of future interest.
Collapse
Affiliation(s)
- Matthew R Lynch
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| | - Mei T Tran
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| | - Samir M Parikh
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
48
|
He L, Wei Q, Liu J, Yi M, Liu Y, Liu H, Sun L, Peng Y, Liu F, Venkatachalam MA, Dong Z. AKI on CKD: heightened injury, suppressed repair, and the underlying mechanisms. Kidney Int 2017; 92:1071-1083. [PMID: 28890325 DOI: 10.1016/j.kint.2017.06.030] [Citation(s) in RCA: 268] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected. Although AKI-to-CKD transition has been intensively studied, the information of AKI on CKD is very limited. Nonetheless, AKI, when occurring in patients with CKD, is known to be more severe and difficult to recover. CKD is associated with significant changes in cell signaling in kidney tissues, including the activation of transforming growth factor-β, p53, hypoxia-inducible factor, and major developmental pathways. At the cellular level, CKD is characterized by mitochondrial dysfunction, oxidative stress, and aberrant autophagy. At the tissue level, CKD is characterized by chronic inflammation and vascular dysfunction. These pathologic changes may contribute to the heightened sensitivity of, and nonrecovery from, AKI in patients with CKD.
Collapse
Affiliation(s)
- Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Jing Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Mixuan Yi
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youming Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Manjeri A Venkatachalam
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA.
| |
Collapse
|
49
|
Delayed Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Inhibition by Trametinib Attenuates Systemic Inflammatory Responses and Multiple Organ Injury in Murine Sepsis. Crit Care Med 2017; 44:e711-20. [PMID: 27031380 DOI: 10.1097/ccm.0000000000001672] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The mitogen-activated protein kinase/extracellular signal-regulated kinase signaling pathway is an essential component of innate immunity necessary for mediating proinflammatory responses in the setting of sepsis. We previously demonstrated that the mitogen-activated protein kinase 1/2 inhibitor trametinib prevents endotoxin-induced renal injury in mice. We therefore assessed efficacy of trametinib in a more clinically relevant experimental model of sepsis. DESIGN Controlled in vivo laboratory study. SETTING University animal research laboratory. SUBJECTS Male C57BL/6 mice. INTERVENTIONS Mice were subjected to cecal ligation and puncture to induce sepsis or underwent sham operation as controls. Six hours after cecal ligation and puncture, mice were randomized to four experimental groups as follows: 1) sham control; 2) sham control + trametinib (1 mg/kg, IP); 3) cecal ligation and puncture; and 4) cecal ligation and puncture + trametinib. All animals received buprenorphine (0.05 mg/kg, SC) and imipenem/cilastatin (14 mg/kg, SC) in 1.5 mL of warm saline (40 mL/kg) at the 6-hour time point. Mice were euthanized at 18 hours after induction of cecal ligation and puncture. MEASUREMENTS AND MAIN RESULTS Trametinib inhibition of mitogen-activated protein kinase/extracellular signal-regulated kinase signaling 6 hours after cecal ligation and puncture attenuated increases in circulating proinflammatory cytokines (tumor necrosis factor-α, interleukin-1β, interleukin-6, and granulocyte macrophage colony-stimulating factor) and hypothermia at 18 hours. Trametinib also attenuated multiple organ injury as determined by serum creatinine, alanine aminotransferase, lactate dehydrogenase, and creatine kinase. At the organ level, trametinib completely restored peritubular capillary perfusion in the kidney. Restoration of microvascular perfusion was associated with reduced messenger RNA expression of well-characterized markers of proximal tubule injury. mitogen-activated protein kinase/extracellular signal-regulated kinase blockade attenuated cecal ligation and puncture-mediated up-regulation of cytokines (tumor necrosis factor-α, interleukin-1β) and restored interleukin-6 to control levels in the renal cortex, indicating the protective effects on the proximal tubule occur primarily through modulation of the proinflammatory response in sepsis. CONCLUSIONS These data reveal that the mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor trametinib attenuates systemic inflammation and multiple organ damage in a clinically relevant model of sepsis. Because trametinib has been safely used in humans, we propose that this drug might represent a translatable approach to limit organ injury in septic patients.
Collapse
|
50
|
Anderberg SB, Luther T, Frithiof R. Physiological aspects of Toll-like receptor 4 activation in sepsis-induced acute kidney injury. Acta Physiol (Oxf) 2017; 219:573-588. [PMID: 27602552 PMCID: PMC5324638 DOI: 10.1111/apha.12798] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/08/2016] [Accepted: 09/05/2016] [Indexed: 12/21/2022]
Abstract
Sepsis‐induced acute kidney injury (SI‐AKI) is common and associated with high mortality. Survivors are at increased risk of chronic kidney disease. The precise mechanism underlying SI‐AKI is unknown, and no curative treatment exists. Toll‐like receptor 4 (TLR4) activates the innate immune system in response to exogenous microbial products. The result is an inflammatory reaction aimed at clearing a potential infection. However, the consequence may also be organ dysfunction as the immune response can cause collateral damage to host tissue. The purpose of this review is to describe the basis for how ligand binding to TLR4 has the potential to cause renal dysfunction and the mechanisms by which this may take place in gram‐negative sepsis. In addition, we highlight areas for future research that can further our knowledge of the pathogenesis of SI‐AKI in relation to TLR4 activation. TLR4 is expressed in the kidney. Activation of TLR4 causes cytokine and chemokine release as well as renal leucocyte infiltration. It also results in endothelial and tubular dysfunction in addition to altered renal metabolism and circulation. From a physiological standpoint, inhibiting TLR4 in large animal experimental SI‐AKI significantly improves renal function. Thus, current evidence indicates that TLR4 has the ability to mediate SI‐AKI by a number of mechanisms. The strong experimental evidence supporting a role of TLR4 in the pathogenesis of SI‐AKI in combination with the availability of pharmacological tools to target TLR4 warrants future human studies.
Collapse
Affiliation(s)
- S. B. Anderberg
- Department of Surgical Sciences; Section of Anesthesia & Intensive Care; Uppsala University; Uppsala Sweden
| | - T. Luther
- Department of Surgical Sciences; Section of Anesthesia & Intensive Care; Uppsala University; Uppsala Sweden
| | - R. Frithiof
- Department of Surgical Sciences; Section of Anesthesia & Intensive Care; Uppsala University; Uppsala Sweden
| |
Collapse
|