1
|
Shi L, Zha H, Zhao J, An H, Huang H, Xia Y, Yan Z, Song Z, Zhu J. Caloric restriction exacerbates renal post-ischemic injury and fibrosis by modulating mTORC1 signaling and autophagy. Redox Biol 2025; 80:103500. [PMID: 39837191 PMCID: PMC11787690 DOI: 10.1016/j.redox.2025.103500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVE This study investigates the effects of caloric restriction (CR) on renal injury and fibrosis following ischemia-reperfusion injury (IRI), with a focus on the roles of the mechanistic/mammalian target of rapamycin complex 1 (mTORC1) signaling and autophagy. METHODS A mouse model of unilateral IRI with or without CR was used. Renal function was assessed through serum creatinine and blood urea nitrogen levels, while histological analysis and molecular assays evaluated tubular injury, fibrosis, mTORC1 signaling, and autophagy activation. Inducible renal tubule-specific Atg7 knockout mice and autophagy inhibitor 3-MA were used to elucidate autophagy's role in renal outcomes. RESULTS CR exacerbated renal dysfunction, tubular injury, and fibrosis in IRI mice, associated with suppressed mTORC1 signaling and enhanced autophagy. Rapamycin, an mTORC1 inhibitor, mimicked the effects of CR, further supporting the involvement of mTORC1-autophagy pathway. Tubule-specific Atg7 knockout and autophagy inhibitor 3-MA mitigated these effects, indicating a central role for autophagy in CR-induced renal damage. Glucose supplementation, but not branched-chain amino acids (BCAAs), alleviated CR-induced renal fibrosis and dysfunction by restoring mTORC1 activation. Finally, we identified leucyl-tRNA synthetase 1 (LARS1) as a key mediator of nutrient sensing and mTORC1 activation, demonstrating its glucose dependency under CR conditions. CONCLUSION Our study provides novel insights into the interplay between nutrient metabolism, mTORC1 signaling, and autophagy in IRI-induced renal damages, offering potential therapeutic targets for mitigating CR-associated complications after renal IRI.
Collapse
Affiliation(s)
- Lang Shi
- Department of Nephrology, The First Hospital of Lanzhou University, Lanzhou, 730000, China; The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Hongchu Zha
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, 443000, China
| | - Juan Zhao
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Haiqian An
- Department of Nephrology, The First Hospital of Lanzhou University, Lanzhou, 730000, China; The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Hua Huang
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, 443000, China
| | - Yao Xia
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, 443000, China
| | - Ziyu Yan
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, 443000, China
| | - Zhixia Song
- Department of Nephrology, The People's Hospital of Longhua, Shenzhen, 518109, China
| | - Jiefu Zhu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
2
|
DiMartino S, Revelo MP, Mallipattu SK, Piret SE. Activation of branched chain amino acid catabolism protects against nephrotoxic acute kidney injury. Am J Physiol Renal Physiol 2025; 328:F152-F163. [PMID: 39653371 PMCID: PMC11918290 DOI: 10.1152/ajprenal.00260.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Acute kidney injury (AKI) is a major risk factor for chronic kidney disease (CKD), and there are currently no therapies for AKI. Proximal tubules (PTs) are particularly susceptible to AKI, due to nephrotoxins such as aristolochic acid I (AAI). Normal PTs use fatty acid oxidation and branched chain amino acid (BCAA; valine, leucine, and isoleucine) catabolism to generate ATP; however, in AKI, these pathways are downregulated. Our aim was to investigate the utility of a pharmacological activator of BCAA catabolism, BT2, in preventing nephrotoxic AKI. Mice were administered two injections of AAI 3 days apart to induce AKI, with or without daily BT2 treatment. Mice treated with BT2 had significantly protected kidney function (reduced serum creatinine and urea nitrogen), reduced histological injury, preservation of PT (Lotus lectin staining), and less PT injury (cytokeratin-20 staining) and inflammatory gene expression compared with mice with AAI alone. Mice with AKI had increased circulating BCAA and accumulation of BCAA in the kidney cortex. Leucine is a potent activator of the mechanistic target of rapamycin complex 1 (mTORC1) signaling, and mTORC1 signaling was activated in mice treated with AAI. However, BT2 reduced kidney cortical BCAA accumulation and attenuated the mTORC1 signaling. In vitro, injured primary PT cells had compromised mitochondrial bioenergetics, but cells treated with AAI + BT2 had partially restored mitochondrial bioenergetics and improved injury markers compared with cells treated with AAI alone. Thus, pharmacological activation of BCAA catabolism using BT2 attenuated nephrotoxic AKI in mice.NEW & NOTEWORTHY This study explored the effects of pharmacological activation of branched chain amino acid (BCAA) catabolism using BT2 to prevent nephrotoxic acute kidney injury (AKI) in mice. Our results indicate that activation of BCAA catabolism protects against nephrotoxic AKI, in association with reduced BCAA accumulation, reduced mammalian target of rapamycin protein complex 1 signaling, and improved mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Samaneh DiMartino
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Monica P Revelo
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States
| | - Sandeep K Mallipattu
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
- Renal Section, Northport VA Medical Center, Northport, New York, United States
| | - Sian E Piret
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| |
Collapse
|
3
|
Grahammer F, Dumoulin B, Gulieva RE, Wu H, Xu Y, Sulaimanov N, Arnold F, Sandner L, Cordts T, Todkar A, Moulin P, Reichardt W, Puelles VG, Kramann R, Freedman BS, Busch H, Boerries M, Walz G, Huber TB. Cyclin-dependent kinase 4 drives cystic kidney disease in the absence of mTORC1 signaling activity. Kidney Int 2024; 106:856-869. [PMID: 39218392 DOI: 10.1016/j.kint.2024.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Progression of cystic kidney disease has been linked to activation of the mTORC1 signaling pathway. Yet the utility of mTORC1 inhibitors to treat patients with polycystic kidney disease remains controversial despite promising preclinical data. To define the cell intrinsic role of mTORC1 for cyst development, the mTORC1 subunit gene Raptor was selectively inactivated in kidney tubular cells lacking cilia due to simultaneous deletion of the kinesin family member gene Kif3A. In contrast to a rapid onset of cyst formation and kidney failure in mice with defective ciliogenesis, both kidney function, cyst formation discerned by magnetic resonance imaging and overall survival were strikingly improved in mice additionally lacking Raptor. However, these mice eventually succumbed to cystic kidney disease despite mTORC1 inactivation. In-depth transcriptome analysis revealed the rapid activation of other growth-promoting signaling pathways, overriding the effects of mTORC1 deletion and identified cyclin-dependent kinase (CDK) 4 as an alternate driver of cyst growth. Additional inhibition of CDK4-dependent signaling by the CDK4/6 inhibitor Palbociclib markedly slowed disease progression in mice and human organoid models of polycystic kidney disease and potentiated the effects of mTORC1 deletion/inhibition. Our findings indicate that cystic kidneys rapidly adopt bypass mechanisms typically observed in drug resistant cancers. Thus, future clinical trials need to consider combinatorial or sequential therapies to improve therapeutic efficacy in patients with cystic kidney disease.
Collapse
Affiliation(s)
- Florian Grahammer
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Bernhard Dumoulin
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ramila E Gulieva
- Division of Nephrology, University of Washington School of Medicine, Seattle, Washington, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, Washington, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Hui Wu
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yaoxian Xu
- Institute of Experimental Medicine and Systems Biology, Medical Faculty, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Nurgazy Sulaimanov
- Department of Electrical Engineering and Information Technology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Frederic Arnold
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Lukas Sandner
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Tomke Cordts
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Abhijeet Todkar
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Pierre Moulin
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | - Wilfried Reichardt
- Department of Diagnostic and Interventional Radiology, Division of Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, Medical Faculty, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Benjamin S Freedman
- Division of Nephrology, University of Washington School of Medicine, Seattle, Washington, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, Washington, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Plurexa LLC, Seattle, Washington, USA
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between Deutsches Krebs Forschungs Zentrum (DKFZ) and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Gerd Walz
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany; Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Swaroop V, Ozkan E, Herrmann L, Thurman A, Kopasz-Gemmen O, Kunamneni A, Inoki K. mTORC1 signaling and diabetic kidney disease. Diabetol Int 2024; 15:707-718. [PMID: 39469564 PMCID: PMC11512951 DOI: 10.1007/s13340-024-00738-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/26/2024] [Indexed: 10/30/2024]
Abstract
Diabetic kidney disease (DKD) represents the most lethal complication in both type 1 and type 2 diabetes. The disease progresses without obvious symptoms and is often refractory when apparent symptoms have emerged. Although the molecular mechanisms underlying the onset/progression of DKD have been extensively studied, only a few effective therapies are currently available. Pathogenesis of DKD involves multifaced events caused by diabetes, which include alterations of metabolisms, signals, and hemodynamics. While the considerable efficacy of sodium/glucose cotransporter-2 (SGLT2) inhibitors or angiotensin II receptor blockers (ARBs) for DKD has been recognized, the ever-increasing number of patients with diabetes and DKD warrants additional practical therapeutic approaches that prevent DKD from diabetes. One plausible but promising target is the mechanistic target of the rapamycin complex 1 (mTORC1) signaling pathway, which senses cellular nutrients to control various anabolic and catabolic processes. This review introduces the current understanding of the mTOR signaling pathway and its roles in the development of DKD and other chronic kidney diseases (CKDs), and discusses potential therapeutic approaches targeting this pathway for the future treatment of DKD.
Collapse
Affiliation(s)
- Vinamra Swaroop
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Eden Ozkan
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Lydia Herrmann
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Aaron Thurman
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | | | | | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, USA
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, USA
| |
Collapse
|
5
|
Demko J, Weber R, Pearce D, Saha B. Aldosterone-independent regulation of K + secretion in the distal nephron. Curr Opin Nephrol Hypertens 2024; 33:526-534. [PMID: 38888034 PMCID: PMC11290980 DOI: 10.1097/mnh.0000000000001006] [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] [Indexed: 06/20/2024]
Abstract
PURPOSE OF REVIEW Maintenance of plasma K + concentration within a narrow range is critical to all cellular functions. The kidneys are the central organ for K + excretion, and robust renal excretory responses to dietary K + loads are essential for survival. Recent advances in the field have challenged the view that aldosterone is at the center of K + regulation. This review will examine recent findings and propose a new mechanism for regulating K + secretion. RECENT FINDINGS Local aldosterone-independent response systems in the distal nephron are increasingly recognized as key components of the rapid response to an acute K + load, as well as playing an essential role in sustained responses to increased dietary K + . The master kinase mTOR, best known for its role in mediating the effects of growth factors and insulin on growth and cellular metabolism, is central to these aldosterone-independent responses. Recent studies have shown that mTOR, particularly in the context of the "type 2" complex (mTORC2), is regulated by K + in a cell-autonomous fashion. SUMMARY New concepts related to cell-autonomous K + signaling and how it interfaces with aldosterone-dependent regulation are emerging. The underlying signaling pathways and effectors of regulated K + secretion, as well as implications for the aldosterone paradox and disease pathogenesis are discussed.
Collapse
Affiliation(s)
- John Demko
- Department of Medicine, Division of Nephrology, University of California at San Francisco, San Francisco, CA, USA
| | - Robert Weber
- Division of Endocrinology, University of California at San Francisco, San Francisco, CA, USA
| | - David Pearce
- Department of Medicine, Division of Nephrology, University of California at San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, CA, USA
| | - Bidisha Saha
- Department of Medicine, Division of Nephrology, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
6
|
Li X, Zhang Y, Chen H, Wu Y, Chen Y, Gong S, Liu Y, Liu H. Inhibition of TFEB deacetylation in proximal tubular epithelial cells (TECs) promotes TFEB activation and alleviates TEC damage in diabetic kidney disease. FASEB J 2024; 38:e23884. [PMID: 39135512 DOI: 10.1096/fj.202302634r] [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: 12/20/2023] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 09/26/2024]
Abstract
The inhibition of the autophagolysosomal pathway mediated by transcription factor EB (TFEB) inactivation in proximal tubular epithelial cells (TECs) is a key mechanism of TEC injury in diabetic kidney disease (DKD). Acetylation is a novel mechanism that regulates TFEB activity. However, there are currently no studies on whether the adjustment of the acetylation level of TFEB can reduce the damage of diabetic TECs. In this study, we investigated the effect of Trichostatin A (TSA), a typical deacetylase inhibitor, on TFEB activity and damage to TECs in both in vivo and in vitro models of DKD. Here, we show that TSA treatment can alleviate the pathological damage of glomeruli and renal tubules and delay the DKD progression in db/db mice, which is associated with the increased expression of TFEB and its downstream genes. In vitro studies further confirmed that TSA treatment can upregulate the acetylation level of TFEB, promote its nuclear translocation, and activate the expression of its downstream genes, thereby reducing the apoptosis level of TECs. TFEB deletion or HDAC6 knockdown in TECs can counteract the activation effect of TSA on autophagolysosomal pathway. We also found that TFEB enhances the transcription of Tfeb through binding to its promoter and promotes its own expression. Our results, thus, provide a novel therapeutic mechanism for DKD that the alleviation of TEC damage by activating the autophagic lysosomal pathway through upregulating TFEB acetylation can, thus, delay DKD progression.
Collapse
Affiliation(s)
- Xiaoyu Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Department of Nephrology, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yaozhi Zhang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Department of Nephrology, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huixia Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Department of Nephrology, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yang Wu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Department of Nephrology, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yongming Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Department of Nephrology, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Siqiao Gong
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Department of Nephrology, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yonghan Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Department of Nephrology, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huafeng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Department of Nephrology, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
7
|
Kaur H, Kamboj K, Naik S, Kumar V, Yadav AK. A pilot study on the differential urine proteomic profile of subjects with community-acquired acute kidney injury who recover versus those who do not recover completely at 4 months after hospital discharge. Front Med (Lausanne) 2024; 11:1412561. [PMID: 39219798 PMCID: PMC7616407 DOI: 10.3389/fmed.2024.1412561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Background Community-acquired acute kidney injury (CA-AKI) is a sudden structural damage and loss of kidney function in otherwise healthy individuals outside of hospital settings having high morbidity and mortality rates worldwide. Long-term sequelae of AKI involve an associated risk of progression to chronic kidney disease (CKD). Serum creatinine (SCr), the currently used clinical parameter for diagnosing AKI, varies greatly with age, gender, diet, and muscle mass. In the present study, we investigated the difference in urinary proteomic profile of subjects that recovered (R) and incompletely recovered (IR) from CA-AKI, 4 months after hospital discharge. Methods Study subjects were recruited from ongoing study of CA-AKI cohort. Patients with either sex or age > 18 years with no underline CKD were enrolled at the time of hospital discharge. Incomplete recovery from CA-AKI was defined as eGFR < 60 mL/min/1.73 m2 or dialysis dependence at 4 months after discharge. Second-morning urine samples were collected, and proteome analysis was performed with LC-MS/MS. Data were analyzed by Proteome Discoverer platform 2.2 (Thermo Scientific) using statistical and various bioinformatics tools for abundance of protein, cellular component, protein class and biological process were analyzed in the recovered and incompletely recovered groups. Results A total of 28 subjects (14 in each group) were enrolled. Collectively, 2019 peptides and proteins with 30 high-abundance proteins in the incompletely recovered group (R/IR <0.5, abundance ratio adj. p-value <0.05) and 11 high-abundance proteins in the incompletely recovered group (R/IR >2.0, abundance ratio adj. p-value <0.05) were identified. Tissue specificity analysis, GO enrichment analysis, and pathway enrichment analysis revealed significant proteins in both the groups that are part of different pathways and might be playing crucial role in renal recovery during the 4-month span after hospital discharge. Conclusion In conclusion, this study helped in identifying potential proteins and associated pathways that are either upregulated or downregulated at the time of hospital discharge in incompletely recovered CA-AKI patients that can be further investigated to check for their exact role in the disease progression or repair.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Experimental Medicine & Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kajal Kamboj
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sachin Naik
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vivek Kumar
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashok Kumar Yadav
- Department of Experimental Medicine & Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
8
|
Wang H, Li X, Zhang Q, Fu C, Jiang W, Xue J, Liu S, Meng Q, Ai L, Zhi X, Deng S, Liang W. Autophagy in Disease Onset and Progression. Aging Dis 2024; 15:1646-1671. [PMID: 37962467 PMCID: PMC11272186 DOI: 10.14336/ad.2023.0815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/15/2023] [Indexed: 11/15/2023] Open
Abstract
Autophagy is a biological phenomenon whereby components of cells can self-degrade using autophagosomes. During this process, cells can clear dysfunctional organelles or unwanted elements. Autophagy can recycle unnecessary biomolecules into new components or sometimes, even destroy the cells themselves. This cellular process was first observed in 1962 by Keith R. Porter et al. Since then, autophagy has been studied for over 60 years, and much has been learned on the topic. Nevertheless, the process is still not fully understood. It has been proven, for example, that autophagy can be a positive force for maintaining good health by removing older or damaged cells. By contrast, autophagy is also involved in the onset and progression of various conditions caused by pathogenic infections. These diseases generally involve several important organs in the human body, including the liver, kidney, heart, and central nervous system. The regulation of the defects of autophagy defects may potentially be used to treat some diseases. This review comprehensively discusses recent research frontiers and topics of interest regarding autophagy-related diseases.
Collapse
Affiliation(s)
- Hao Wang
- Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, Guangdong, China.
| | - Xiushen Li
- Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China.
| | - Qi Zhang
- Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China.
| | - Chengtao Fu
- School of Medicine, Huzhou University, Zhejiang, China.
| | - Wenjie Jiang
- Department of Artificial Intelligence and Data Science, Hebei University of Technology, Tianjin, China.
| | - Jun Xue
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Shan Liu
- Bioimaging Core of Shenzhen Bay Laboratory Shenzhen, China.
| | - Qingxue Meng
- Technology Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Lisha Ai
- Department of Teaching and Research, Shenzhen University General Hospital, Shenzhen, Guangdong, China.
| | - Xuejun Zhi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Shoulong Deng
- National Health Commission of China (NHC) Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China.
| | - Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| |
Collapse
|
9
|
Das F, Ghosh-Choudhury N, Kasinath BS, Sharma K, Choudhury GG. High glucose-induced downregulation of PTEN-Long is sufficient for proximal tubular cell injury in diabetic kidney disease. Exp Cell Res 2024; 440:114116. [PMID: 38830568 DOI: 10.1016/j.yexcr.2024.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 04/24/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
During the progression of diabetic kidney disease, proximal tubular epithelial cells respond to high glucose to induce hypertrophy and matrix expansion leading to renal fibrosis. Recently, a non-canonical PTEN has been shown to be translated from an upstream initiation codon CUG (leucine) to produce a longer protein called PTEN-Long (PTEN-L). Interestingly, the extended sequence present in PTEN-L contains cell secretion/penetration signal. Role of this non-canonical PTEN-L in diabetic renal tubular injury is not known. We show that high glucose decreases expression of PTEN-L. As a mechanism of its function, we find that reduced PTEN-L activates Akt-2, which phosphorylates and inactivate tuberin and PRAS40, resulting in activation of mTORC1 in tubular cells. Antibacterial agent acriflavine and antiviral agent ATA regulate translation from CUG codon. Acriflavine and ATA, respectively, decreased and increased expression of PTEN-L to altering Akt-2 and mTORC1 activation in the absence of change in expression of canonical PTEN. Consequently, acriflavine and ATA modulated high glucose-induced tubular cell hypertrophy and lamininγ1 expression. Importantly, expression of PTEN-L inhibited high glucose-stimulated Akt/mTORC1 activity to abrogate these processes. Since PTEN-L contains secretion/penetration signals, addition of conditioned medium containing PTEN-L blocked Akt-2/mTORC1 activity. Notably, in renal cortex of diabetic mice, we found reduced PTEN-L concomitant with Akt-2/mTORC1 activation, leading to renal hypertrophy and lamininγ1 expression. These results present first evidence for involvement of PTEN-L in diabetic kidney disease.
Collapse
Affiliation(s)
- Falguni Das
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA
| | | | | | - Kumar Sharma
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA
| | - Goutam Ghosh Choudhury
- VA Research, South Texas Veterans Health Care System, San Antonio, TX, USA; Department of Medicine, TX, USA; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA.
| |
Collapse
|
10
|
deRiso J, Mukherjee M, Janga M, Simmons A, Kareta M, Tao J, Chandrasekar I, Surendran K. Kidney collecting duct cell type composition is regulated by Notch signaling via modulation of mTORC1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.587573. [PMID: 38645025 PMCID: PMC11030444 DOI: 10.1101/2024.04.09.587573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The plasticity and diversity of cell types with specialized functions likely defines the capacity of multicellular organisms to adapt to physiologic stressors. The kidney collecting ducts contribute to water, electrolyte, and pH homeostasis and are composed of mature intermingled epithelial cell types that are susceptible to transdifferentiate. The conversion of kidney collecting duct principal cells to intercalated cells is actively inhibited by Notch signaling to ensure urine concentrating capability. Here we identify Hes1, a target of Notch signaling, allows for maintenance of functionally distinct epithelial cell types within the same microenvironment by regulating mechanistic target of rapamycin complex 1 (mTORC1) activity. Hes1 directly represses the expression of insulin receptor substrate 1 ( Irs1 ), an upstream component of mTOR pathway and suppresses mTORC1 activity in principal cells. Genetic inactivation of tuberous sclerosis complex 2 ( Tsc2 ) to increase mTORC1 activity in mature principal cells is sufficient to promote acquisition of intercalated cell properties, while inhibition of mTORC1 in adult kidney epithelia suppresses intercalated cell properties. Considering that mTORC1 integrates environmental cues, the linkage of functionally distinct epithelial cell types to mTORC1 activity levels likely allows for cell plasticity to be regulated by physiologic and metabolic signals and the ability to sense/transduce these signals.
Collapse
|
11
|
Chrysopoulou M, Rinschen MM. Metabolic Rewiring and Communication: An Integrative View of Kidney Proximal Tubule Function. Annu Rev Physiol 2024; 86:405-427. [PMID: 38012048 DOI: 10.1146/annurev-physiol-042222-024724] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The kidney proximal tubule is a key organ for human metabolism. The kidney responds to stress with altered metabolite transformation and perturbed metabolic pathways, an ultimate cause for kidney disease. Here, we review the proximal tubule's metabolic function through an integrative view of transport, metabolism, and function, and embed it in the context of metabolome-wide data-driven research. Function (filtration, transport, secretion, and reabsorption), metabolite transformation, and metabolite signaling determine kidney metabolic rewiring in disease. Energy metabolism and substrates for key metabolic pathways are orchestrated by metabolite sensors. Given the importance of renal function for the inner milieu, we also review metabolic communication routes with other organs. Exciting research opportunities exist to understand metabolic perturbation of kidney and proximal tubule function, for example, in hypertension-associated kidney disease. We argue that, based on the integrative view outlined here, kidney diseases without genetic cause should be approached scientifically as metabolic diseases.
Collapse
Affiliation(s)
| | - Markus M Rinschen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark;
- III. Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Moellmann J, Krueger K, Wong DWL, Klinkhammer BM, Buhl EM, Dehairs J, Swinnen JV, Noels H, Jankowski J, Lebherz C, Boor P, Marx N, Lehrke M. 2,8-Dihydroxyadenine-induced nephropathy causes hexosylceramide accumulation with increased mTOR signaling, reduced levels of protective SirT3 expression and impaired renal mitochondrial function. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166825. [PMID: 37536502 DOI: 10.1016/j.bbadis.2023.166825] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
AIM Chronic kidney disease (CKD) is accompanied by increased cardiovascular risk and heart failure (HF). In rodents, 2,8-dihydroxyadenine (DHA)-induced nephropathy is a frequently used CKD model. Cardiac and kidney tubular cells share high energy demand to guarantee constant contractive force of the heart or reabsorption/secretion of primary filtrated molecules and waste products by the kidney. Here we analyze time-dependent mechanisms of kidney damage and cardiac consequences under consideration of energetic pathways with the focus on mitochondrial function and lipid metabolism in mice. METHODS AND RESULTS CKD was induced by alternating dietary adenine supplementation (0.2 % or 0.05 % of adenine) in C57BL/6J mice for 9 weeks. Progressive kidney damage led to reduced creatinine clearance, kidney fibrosis and renal inflammation after 3, 6, and 9 weeks. No difference in cardiac function, mitochondrial respiration nor left ventricular fibrosis was observed at any time point. Investigating mechanisms of renal damage, protective SirT3 was decreased in CKD, which contrasted an increase in protein kinase B (AKT) expression, mechanistic target of rapamycin (mTOR) downstream signaling, induction of oxidative and endoplasmic reticulum (ER) stress. This occurred together with impaired renal mitochondrial function and accumulation of hexosylceramides (HexCer) as an established mediator of inflammation and mitochondrial dysfunction in the kidney. CONCLUSIONS 2,8-DHA-induced CKD results in renal activation of the mTOR downstream signaling, endoplasmic reticulum stress, tubular injury, fibrosis, inflammation, oxidative stress and impaired kidney mitochondrial function in conjunction with renal hexosylceramide accumulation in C57BL/6J mice.
Collapse
Affiliation(s)
- Julia Moellmann
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Katja Krueger
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Dickson W L Wong
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Barbara M Klinkhammer
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Eva M Buhl
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany; Department of Nephrology, RWTH Aachen University, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University, Aachen, Germany
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, LKI - Leuven Cancer Institute, KU Leuven - University of Leuven, Leuven, Belgium
| | - Johan V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, LKI - Leuven Cancer Institute, KU Leuven - University of Leuven, Leuven, Belgium
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Corinna Lebherz
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany; Department of Nephrology, RWTH Aachen University, Aachen, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
13
|
Chu LK, Cao X, Wan L, Diao Q, Zhu Y, Kan Y, Ye LL, Mao YM, Dong XQ, Xiong QW, Fu MC, Zhang T, Zhou HT, Cai SZ, Ma ZR, Hsu SW, Wu R, Chen CH, Yan XM, Liu J. Autophagy of OTUD5 destabilizes GPX4 to confer ferroptosis-dependent kidney injury. Nat Commun 2023; 14:8393. [PMID: 38110369 PMCID: PMC10728081 DOI: 10.1038/s41467-023-44228-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
Ferroptosis is an iron-dependent programmed cell death associated with severe kidney diseases, linked to decreased glutathione peroxidase 4 (GPX4). However, the spatial distribution of renal GPX4-mediated ferroptosis and the molecular events causing GPX4 reduction during ischemia-reperfusion (I/R) remain largely unknown. Using spatial transcriptomics, we identify that GPX4 is situated at the interface of the inner cortex and outer medulla, a hyperactive ferroptosis site post-I/R injury. We further discover OTU deubiquitinase 5 (OTUD5) as a GPX4-binding protein that confers ferroptosis resistance by stabilizing GPX4. During I/R, ferroptosis is induced by mTORC1-mediated autophagy, causing OTUD5 degradation and subsequent GPX4 decay. Functionally, OTUD5 deletion intensifies renal tubular cell ferroptosis and exacerbates acute kidney injury, while AAV-mediated OTUD5 delivery mitigates ferroptosis and promotes renal function recovery from I/R injury. Overall, this study highlights a new autophagy-dependent ferroptosis module: hypoxia/ischemia-induced OTUD5 autophagy triggers GPX4 degradation, offering a potential therapeutic avenue for I/R-related kidney diseases.
Collapse
Affiliation(s)
- Li-Kai Chu
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Xu Cao
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Lin Wan
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Qiang Diao
- Department of Medical Imaging, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 210002, Nanjing, China
| | - Yu Zhu
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Yu Kan
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Li-Li Ye
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Yi-Ming Mao
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, 215028, Suzhou, China
| | - Xing-Qiang Dong
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Qian-Wei Xiong
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Ming-Cui Fu
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Ting Zhang
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Hui-Ting Zhou
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Shi-Zhong Cai
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Zhou-Rui Ma
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China
| | - Ssu-Wei Hsu
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Reen Wu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Ching-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, USA.
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA.
| | - Xiang-Ming Yan
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China.
| | - Jun Liu
- Pediatric Institute of Soochow University, Children's Hospital of Soochow University, Soochow University, 215025, Suzhou, China.
| |
Collapse
|
14
|
Wu Q, Huang F. LncRNA H19: a novel player in the regulation of diabetic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1238981. [PMID: 37964955 PMCID: PMC10641825 DOI: 10.3389/fendo.2023.1238981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Diabetic kidney disease (DKD), one of the most severe complications of diabetes mellitus (DM), has received considerable attention owing to its increasing prevalence and contribution to chronic kidney disease (CKD) and end-stage kidney disease (ESRD). However, the use of drugs targeting DKD remains limited. Recent data suggest that long non-coding RNAs (lncRNAs) play a vital role in the development of DKD. The lncRNA H19 is the first imprinted gene, which is expressed in the embryo and down-regulated at birth, and its role in tumors has long been a subject of controversy, however, in recent years, it has received increasing attention in kidney disease. The LncRNA H19 is engaged in the pathological progression of DKD, including glomerulosclerosis and tubulointerstitial fibrosis via the induction of inflammatory responses, apoptosis, ferroptosis, pyroptosis, autophagy, and oxidative damage. In this review, we highlight the most recent research on the molecular mechanism and regulatory forms of lncRNA H19 in DKD, including epigenetic, post-transcriptional, and post-translational regulation, providing a new predictive marker and therapeutic target for the management of DKD.
Collapse
Affiliation(s)
| | - Fengjuan Huang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Nasrah S, Radi A, Daberkow JK, Hummler H, Weber S, Seaayfan E, Kömhoff M. MAGED2 Depletion Promotes Stress-Induced Autophagy by Impairing the cAMP/PKA Pathway. Int J Mol Sci 2023; 24:13433. [PMID: 37686237 PMCID: PMC10488052 DOI: 10.3390/ijms241713433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Melanoma-associated antigen D2 (MAGED2) plays an essential role in activating the cAMP/PKA pathway under hypoxic conditions, which is crucial for stimulating renal salt reabsorption and thus explaining the transient variant of Bartter's syndrome. The cAMP/PKA pathway is also known to regulate autophagy, a lysosomal degradation process induced by cellular stress. Previous studies showed that two members of the melanoma-associated antigens MAGE-family inhibit autophagy. To explore the potential role of MAGED2 in stress-induced autophagy, specific MAGED2-siRNA were used in HEK293 cells under physical hypoxia and oxidative stress (cobalt chloride, hypoxia mimetic). Depletion of MAGED2 resulted in reduced p62 levels and upregulation of both the autophagy-related genes (ATG5 and ATG12) as well as the autophagosome marker LC3II compared to control siRNA. The increase in the autophagy markers in MAGED2-depleted cells was further confirmed by leupeptin-based assay which concurred with the highest LC3II accumulation. Likewise, under hypoxia, immunofluorescence in HEK293, HeLa and U2OS cell lines demonstrated a pronounced accumulation of LC3B puncta upon MAGED2 depletion. Moreover, LC3B puncta were absent in human fetal control kidneys but markedly expressed in a fetal kidney from a MAGED2-deficient subject. Induction of autophagy with both physical hypoxia and oxidative stress suggests a potentially general role of MAGED2 under stress conditions. Various other cellular stressors (brefeldin A, tunicamycin, 2-deoxy-D-glucose, and camptothecin) were analyzed, which all induced autophagy in the absence of MAGED2. Forskolin (FSK) inhibited, whereas GNAS Knockdown induced autophagy under hypoxia. In contrast to other MAGE proteins, MAGED2 has an inhibitory role on autophagy only under stress conditions. Hence, a prominent role of MAGED2 in the regulation of autophagy under stress conditions is evident, which may also contribute to impaired fetal renal salt reabsorption by promoting autophagy of salt-transporters in patients with MAGED2 mutation.
Collapse
Affiliation(s)
- Sadiq Nasrah
- Department of Pediatrics, University Hospital Giessen and Marburg, Philipps University Marburg, 35043 Marburg, Germany; (S.N.); (A.R.); (H.H.); (S.W.)
| | - Aline Radi
- Department of Pediatrics, University Hospital Giessen and Marburg, Philipps University Marburg, 35043 Marburg, Germany; (S.N.); (A.R.); (H.H.); (S.W.)
| | - Johanna K. Daberkow
- Faculty of Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Helmut Hummler
- Department of Pediatrics, University Hospital Giessen and Marburg, Philipps University Marburg, 35043 Marburg, Germany; (S.N.); (A.R.); (H.H.); (S.W.)
| | - Stefanie Weber
- Department of Pediatrics, University Hospital Giessen and Marburg, Philipps University Marburg, 35043 Marburg, Germany; (S.N.); (A.R.); (H.H.); (S.W.)
| | - Elie Seaayfan
- Department of Pediatrics, University Hospital Giessen and Marburg, Philipps University Marburg, 35043 Marburg, Germany; (S.N.); (A.R.); (H.H.); (S.W.)
| | - Martin Kömhoff
- Department of Pediatrics, University Hospital Giessen and Marburg, Philipps University Marburg, 35043 Marburg, Germany; (S.N.); (A.R.); (H.H.); (S.W.)
| |
Collapse
|
16
|
In Humanized Sickle Cell Mice, Imatinib Protects Against Sickle Cell-Related Injury. Hemasphere 2023; 7:e848. [PMID: 36874380 PMCID: PMC9977487 DOI: 10.1097/hs9.0000000000000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/19/2023] [Indexed: 03/06/2023] Open
Abstract
Drug repurposing is a valuable strategy for rare diseases. Sickle cell disease (SCD) is a rare hereditary hemolytic anemia accompanied by acute and chronic painful episodes, most often in the context of vaso-occlusive crisis (VOC). Although progress in the knowledge of pathophysiology of SCD have allowed the development of new therapeutic options, a large fraction of patients still exhibits unmet therapeutic needs, with persistence of VOCs and chronic disease progression. Here, we show that imatinib, an oral tyrosine kinase inhibitor developed for the treatment of chronic myelogenous leukemia, acts as multimodal therapy targeting signal transduction pathways involved in the pathogenesis of both anemia and inflammatory vasculopathy of humanized murine model for SCD. In addition, imatinib inhibits the platelet-derived growth factor-B-dependent pathway, interfering with the profibrotic response to hypoxia/reperfusion injury, used to mimic acute VOCs. Our data indicate that imatinib might be considered as possible new therapeutic tool for chronic treatment of SCD.
Collapse
|
17
|
Huynh C, Ryu J, Lee J, Inoki A, Inoki K. Nutrient-sensing mTORC1 and AMPK pathways in chronic kidney diseases. Nat Rev Nephrol 2023; 19:102-122. [PMID: 36434160 DOI: 10.1038/s41581-022-00648-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
Nutrients such as glucose, amino acids and lipids are fundamental sources for the maintenance of essential cellular processes and homeostasis in all organisms. The nutrient-sensing kinases mechanistic target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) are expressed in many cell types and have key roles in the control of cell growth, proliferation, differentiation, metabolism and survival, ultimately contributing to the physiological development and functions of various organs, including the kidney. Dysregulation of these kinases leads to many human health problems, including cancer, neurodegenerative diseases, metabolic disorders and kidney diseases. In the kidney, physiological levels of mTOR and AMPK activity are required to support kidney cell growth and differentiation and to maintain kidney cell integrity and normal nephron function, including transport of electrolytes, water and glucose. mTOR forms two functional multi-protein kinase complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Hyperactivation of mTORC1 leads to podocyte and tubular cell dysfunction and vulnerability to injury, thereby contributing to the development of chronic kidney diseases, including diabetic kidney disease, obesity-related kidney disease and polycystic kidney disease. Emerging evidence suggests that targeting mTOR and/or AMPK could be an effective therapeutic approach to controlling or preventing these diseases.
Collapse
Affiliation(s)
- Christopher Huynh
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jaewhee Ryu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jooho Lee
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ayaka Inoki
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Is bariatric surgery improving mitochondrial function in the renal cells of patients with obesity-induced kidney disease? Pharmacol Res 2022; 185:106488. [DOI: 10.1016/j.phrs.2022.106488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/22/2022]
|
19
|
Zhu J, Wang H, Jiang X. mTORC1 beyond anabolic metabolism: Regulation of cell death. J Biophys Biochem Cytol 2022; 221:213609. [PMID: 36282248 PMCID: PMC9606688 DOI: 10.1083/jcb.202208103] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1), a multi-subunit protein kinase complex, interrogates growth factor signaling with cellular nutrient and energy status to control metabolic homeostasis. Activation of mTORC1 promotes biosynthesis of macromolecules, including proteins, lipids, and nucleic acids, and simultaneously suppresses catabolic processes such as lysosomal degradation of self-constituents and extracellular components. Metabolic regulation has emerged as a critical determinant of various cellular death programs, including apoptosis, pyroptosis, and ferroptosis. In this article, we review the expanding knowledge on how mTORC1 coordinates metabolic pathways to impinge on cell death regulation. We focus on the current understanding on how nutrient status and cellular signaling pathways connect mTORC1 activity with ferroptosis, an iron-dependent cell death program that has been implicated in a plethora of human diseases. In-depth understanding of the principles governing the interaction between mTORC1 and cell death pathways can ultimately guide the development of novel therapies for the treatment of relevant pathological conditions.
Collapse
Affiliation(s)
- Jiajun Zhu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China,Tsinghua-Peking Center for Life Sciences, Beijing, China,Correspondence to Jiajun Zhu:
| | - Hua Wang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY,Xuejun Jiang:
| |
Collapse
|
20
|
Spitz D, Comas M, Gerstner L, Kayser S, Helmstädter M, Walz G, Hermle T. mTOR-Dependent Autophagy Regulates Slit Diaphragm Density in Podocyte-like Drosophila Nephrocytes. Cells 2022; 11:2103. [PMID: 35805186 PMCID: PMC9265458 DOI: 10.3390/cells11132103] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Both mTOR signaling and autophagy are important modulators of podocyte homeostasis, regeneration, and aging and have been implicated in glomerular diseases. However, the mechanistic role of these pathways for the glomerular filtration barrier remains poorly understood. We used Drosophila nephrocytes as an established podocyte model and found that inhibition of mTOR signaling resulted in increased spacing between slit diaphragms. Gain-of-function of mTOR signaling did not affect spacing, suggesting that additional cues limit the maximal slit diaphragm density. Interestingly, both activation and inhibition of mTOR signaling led to decreased nephrocyte function, indicating that a fine balance of signaling activity is needed for proper function. Furthermore, mTOR positively controlled cell size, survival, and the extent of the subcortical actin network. We also showed that basal autophagy in nephrocytes is required for survival and limits the expression of the sns (nephrin) but does not directly affect slit diaphragm formation or endocytic activity. However, using a genetic rescue approach, we demonstrated that excessive, mTOR-dependent autophagy is primarily responsible for slit diaphragm misspacing. In conclusion, we established this invertebrate podocyte model for mechanistic studies on the role of mTOR signaling and autophagy, and we discovered a direct mTOR/autophagy-dependent regulation of the slit diaphragm architecture.
Collapse
Affiliation(s)
- Dominik Spitz
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany; (D.S.); (L.G.); (S.K.); (M.H.); (G.W.)
| | - Maria Comas
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany; (D.S.); (L.G.); (S.K.); (M.H.); (G.W.)
| | - Lea Gerstner
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany; (D.S.); (L.G.); (S.K.); (M.H.); (G.W.)
| | - Séverine Kayser
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany; (D.S.); (L.G.); (S.K.); (M.H.); (G.W.)
| | - Martin Helmstädter
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany; (D.S.); (L.G.); (S.K.); (M.H.); (G.W.)
| | - Gerd Walz
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany; (D.S.); (L.G.); (S.K.); (M.H.); (G.W.)
- CIBSS—Centre for Integrative Biological Signalling Studies, 79106 Freiburg, Germany
| | - Tobias Hermle
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, 79106 Freiburg, Germany; (D.S.); (L.G.); (S.K.); (M.H.); (G.W.)
| |
Collapse
|
21
|
Zhu Z, Hu J, Chen Z, Feng J, Yang X, Liang W, Ding G. Transition of acute kidney injury to chronic kidney disease: role of metabolic reprogramming. Metabolism 2022; 131:155194. [PMID: 35346693 DOI: 10.1016/j.metabol.2022.155194] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/04/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is a global public health concern associated with high morbidity and mortality. Although advances in medical management have improved the in-hospital mortality of severe AKI patients, the renal prognosis for AKI patients in the later period is not encouraging. Recent epidemiological investigations have indicated that AKI significantly increases the risk for the development of chronic kidney disease (CKD) and end-stage renal disease (ESRD) in the future, further contributing to the economic burden on health care systems. The transition of AKI to CKD is complex and often involves multiple mechanisms. Recent studies have suggested that renal tubular epithelial cells (TECs) are more prone to metabolic reprogramming during AKI, in which the metabolic process in the TECs shifts from fatty acid β-oxidation (FAO) to glycolysis due to hypoxia, mitochondrial dysfunction, and disordered nutrient-sensing pathways. This change is a double-edged role. On the one hand, enhanced glycolysis acts as a compensation pathway for ATP production; on the other hand, long-term shut down of FAO and enhanced glycolysis lead to inflammation, lipid accumulation, and fibrosis, contributing to the transition of AKI to CKD. This review discusses developments and therapies focused on the metabolic reprogramming of TECs during AKI, and the emerging questions in this evolving field.
Collapse
Affiliation(s)
- Zijing Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Xueyan Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, 430060 Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
22
|
Sako K, Furuichi K, Makiishi S, Yamamura Y, Okumura T, Le T, Kitajima S, Toyama T, Hara A, Iwata Y, Sakai N, Shimizu M, Niimura F, Matsusaka T, Kaneko S, Wada T. Cyclin-dependent kinase 4-related tubular epithelial cell proliferation is regulated by Paired box gene 2 in kidney ischemia-reperfusion injury. Kidney Int 2022; 102:45-57. [PMID: 35483529 DOI: 10.1016/j.kint.2022.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/08/2022] [Accepted: 03/09/2022] [Indexed: 12/20/2022]
Abstract
Paired box 2 (Pax2) is a transcription factor essential for kidney development and is reactivated in proximal tubular epithelial cells (PTECs) during recovery from kidney injury. However, the role of Pax2 in this process is still unknown. Here the role of Pax2 reactivation during injury was examined in the proliferation of PTECs using an ischemia-reperfusion injury (IRI) mouse model. Kidney proximal tubule-specific Pax2 conditional knockout mice were generated by mating kidney androgen-regulated protein-Cre and Pax2 flox mice. The degree of cell proliferation and fibrosis was assessed and a Pax2 inhibitor (EG1) was used to evaluate the role of Pax2 in the hypoxic condition of cultured PTECs (O2 5%, 24 hours). The number of Pax2-positive cells and Pax2 mRNA increased after IRI. Sirius red staining indicated that the area of interstitial fibrosis was significantly larger in knockout mice 14 days after IRI. The number of Ki-67-positive cells (an index of proliferation) was significantly lower in knockout than in wild-type mice after IRI, whereas the number of TUNEL-positive cells (an index of apoptotic cells) was significantly higher in knockout mice four days after IRI. Expression analyses of cell cycle-related genes showed that cyclin-dependent kinase 4 (CDK4) was significantly less expressed in the Pax2 knockout mice. In vitro data showed that the increase in CDK4 mRNA and protein expression induced by hypoxia was attenuated by EG1. Thus, Pax2 reactivation may be involved in PTEC proliferation by activating CDK4, thereby limiting kidney fibrosis.
Collapse
Affiliation(s)
- Keisuke Sako
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kengo Furuichi
- Department of Nephrology, School of Medicine, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Shohei Makiishi
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yuta Yamamura
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Toshiya Okumura
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Thu Le
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Shinji Kitajima
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Akinori Hara
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasunori Iwata
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan; Division of Infection Control, Kanazawa University, Kanazawa, Japan
| | - Norihiko Sakai
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Miho Shimizu
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Fumio Niimura
- Department of Pediatrics, School of Medicine, Tokai University, Isehara, Japan
| | - Taiji Matsusaka
- Department of Basic Medicine, School of Medicine, Tokai University, Isehara, Japan; Institute of Medical Science, Tokai University, Isehara, Japan
| | - Shuichi Kaneko
- Department of System Biology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
23
|
Laouari D, Vergnaud P, Hirose T, Zaidan M, Rabant M, Nguyen C, Burtin M, Legendre C, Codogno P, Friedlander G, Anglicheau D, Terzi F. The sexual dimorphism of kidney growth in mice and humans. Kidney Int 2022; 102:78-95. [PMID: 35337891 DOI: 10.1016/j.kint.2022.02.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 02/02/2022] [Accepted: 02/16/2022] [Indexed: 10/18/2022]
Abstract
Kidney mass and function are sexually determined, but the cellular events and the molecular mechanisms involved in this dimorphism are poorly characterized. By combining female and male mice with castration/replacement experiments, we showed that male mice exhibited kidney overgrowth from five weeks of age. This effect was organ specific, since liver and heart weight were comparable between males and females, regardless of age. Consistently, the androgen receptor was found to be expressed in the kidneys of males, but not in the liver. In growing mice, androgens led to kidney overgrowth by first inducing a burst of cell proliferation and then an increase of cell size. Remarkably, androgens were also required to maintain cell size in adults. In fact, orchiectomy resulted in smaller kidneys in a matter of few weeks. These changes paralleled the changes of the expression of ornithine decarboxylase and cyclin D1, two known mediators of kidney growth, whereas, unexpectedly, mTORC1 and Hippo pathways did not seem to be involved. Androgens also enhanced kidney autophagy, very likely by increasing transcription factor EB nuclear translocation. Functionally, the increase of tubular mass resulted in increased sodium/phosphate transport. These findings were relevant to humans. Remarkably, by studying living gender-paired kidney donors-recipients, we showed that tubular cell size increased three months after transplantation in men as compared to women, regardless of the donor gender. Thus, our results identify novel signaling pathways that may be involved in androgen-induced kidney growth and homeostasis, and suggest that androgens determine kidney size after transplantation.
Collapse
Affiliation(s)
- Denise Laouari
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France
| | - Paul Vergnaud
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France; Service de Néphrologie Pédiatrique-Hémodialyse-Transplantation, AP-HP, Hôpital Necker, Paris, France
| | - Takuo Hirose
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France
| | - Mohamad Zaidan
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France; Service de Néphrologie-Transplantation, AP-HP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Marion Rabant
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France; Service d'Anatomo-Pathologie, AP-HP, Hôpital Necker, Paris, France
| | - Clément Nguyen
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France
| | - Martine Burtin
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France
| | - Christophe Legendre
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France; Service de Néphrologie-Transplantation, AP-HP, Hôpital Necker, Paris, France
| | - Patrice Codogno
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France
| | - Gerard Friedlander
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France
| | - Dany Anglicheau
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France; Service de Néphrologie-Transplantation, AP-HP, Hôpital Necker, Paris, France
| | - Fabiola Terzi
- Université de Paris, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades (INEM), Département « Croissance et Signalisation », F-75006 Paris, France.
| |
Collapse
|
24
|
Jansen J, Reimer KC, Nagai JS, Varghese FS, Overheul GJ, de Beer M, Roverts R, Daviran D, Fermin LA, Willemsen B, Beukenboom M, Djudjaj S, von Stillfried S, van Eijk LE, Mastik M, Bulthuis M, Dunnen WD, van Goor H, Hillebrands JL, Triana SH, Alexandrov T, Timm MC, van den Berge BT, van den Broek M, Nlandu Q, Heijnert J, Bindels EM, Hoogenboezem RM, Mooren F, Kuppe C, Miesen P, Grünberg K, Ijzermans T, Steenbergen EJ, Czogalla J, Schreuder MF, Sommerdijk N, Akiva A, Boor P, Puelles VG, Floege J, Huber TB, van Rij RP, Costa IG, Schneider RK, Smeets B, Kramann R, Achdout H, Aimon A, Bar-David E, Barr H, Ben-Shmuel A, Bennett J, Boby ML, Borden B, Bowman GR, Brun J, BVNBS S, Calmiano M, Carbery A, Cattermole E, Chernychenko E, Choder JD, Clyde A, Coffland JE, Cohen G, Cole J, Contini A, Cox L, Cvitkovic M, Dias A, Donckers K, Dotson DL, Douangamath A, Duberstein S, Dudgeon T, Dunnett L, Eastman PK, Erez N, Eyermann CJ, Fairhead M, Fate G, Fearon D, Federov O, Ferla M, Fernandes RS, Ferrins L, Foster R, Foster H, Gabizon R, Garcia-Sastre A, Gawriljuk VO, Gehrtz P, Gileadi C, Giroud C, Glass WG, Glen R, Itai glinert, Godoy AS, Gorichko M, et alJansen J, Reimer KC, Nagai JS, Varghese FS, Overheul GJ, de Beer M, Roverts R, Daviran D, Fermin LA, Willemsen B, Beukenboom M, Djudjaj S, von Stillfried S, van Eijk LE, Mastik M, Bulthuis M, Dunnen WD, van Goor H, Hillebrands JL, Triana SH, Alexandrov T, Timm MC, van den Berge BT, van den Broek M, Nlandu Q, Heijnert J, Bindels EM, Hoogenboezem RM, Mooren F, Kuppe C, Miesen P, Grünberg K, Ijzermans T, Steenbergen EJ, Czogalla J, Schreuder MF, Sommerdijk N, Akiva A, Boor P, Puelles VG, Floege J, Huber TB, van Rij RP, Costa IG, Schneider RK, Smeets B, Kramann R, Achdout H, Aimon A, Bar-David E, Barr H, Ben-Shmuel A, Bennett J, Boby ML, Borden B, Bowman GR, Brun J, BVNBS S, Calmiano M, Carbery A, Cattermole E, Chernychenko E, Choder JD, Clyde A, Coffland JE, Cohen G, Cole J, Contini A, Cox L, Cvitkovic M, Dias A, Donckers K, Dotson DL, Douangamath A, Duberstein S, Dudgeon T, Dunnett L, Eastman PK, Erez N, Eyermann CJ, Fairhead M, Fate G, Fearon D, Federov O, Ferla M, Fernandes RS, Ferrins L, Foster R, Foster H, Gabizon R, Garcia-Sastre A, Gawriljuk VO, Gehrtz P, Gileadi C, Giroud C, Glass WG, Glen R, Itai glinert, Godoy AS, Gorichko M, Gorrie-Stone T, Griffen EJ, Hart SH, Heer J, Henry M, Hill M, Horrell S, Hurley MF, Israely T, Jajack A, Jnoff E, Jochmans D, John T, De Jonghe S, Kantsadi AL, Kenny PW, Kiappes J, Koekemoer L, Kovar B, Krojer T, Lee AA, Lefker BA, Levy H, London N, Lukacik P, Macdonald HB, Maclean B, Malla TR, Matviiuk T, McCorkindale W, McGovern BL, Melamed S, Michurin O, Mikolajek H, Milne BF, Morris A, Morris GM, Morwitzer MJ, Moustakas D, Nakamura AM, Neto JB, Neyts J, Nguyen L, Noske GD, Oleinikovas V, Oliva G, Overheul GJ, Owen D, Psenak V, Pai R, Pan J, Paran N, Perry B, Pingle M, Pinjari J, Politi B, Powell A, Puni R, Rangel VL, Reddi RN, Reid SP, Resnick E, Ripka EG, Robinson MC, Robinson RP, Rodriguez-Guerra J, Rosales R, Rufa D, Schofield C, Shafeev M, Shaikh A, Shi J, Shurrush K, Sing S, Sittner A, Skyner R, Smalley A, Smilova MD, Solmesky LJ, Spencer J, Strain-Damarell C, Swamy V, Tamir H, Tennant R, Thompson W, Thompson A, Thompson W, Tomasia S, Tumber A, Vakonakis I, van Rij RP, van Geel L, Varghese FS, Vaschetto M, Vitner EB, Voelz V, Volkamer A, von Delft F, von Delft A, Walsh M, Ward W, Weatherall C, Weiss S, White KM, Wild CF, Wittmann M, Wright N, Yahalom-Ronen Y, Zaidmann D, Zidane H, Zitzmann N. SARS-CoV-2 infects the human kidney and drives fibrosis in kidney organoids. Cell Stem Cell 2022; 29:217-231.e8. [PMID: 35032430 PMCID: PMC8709832 DOI: 10.1016/j.stem.2021.12.010] [Show More Authors] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 09/03/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Kidney failure is frequently observed during and after COVID-19, but it remains elusive whether this is a direct effect of the virus. Here, we report that SARS-CoV-2 directly infects kidney cells and is associated with increased tubule-interstitial kidney fibrosis in patient autopsy samples. To study direct effects of the virus on the kidney independent of systemic effects of COVID-19, we infected human-induced pluripotent stem-cell-derived kidney organoids with SARS-CoV-2. Single-cell RNA sequencing indicated injury and dedifferentiation of infected cells with activation of profibrotic signaling pathways. Importantly, SARS-CoV-2 infection also led to increased collagen 1 protein expression in organoids. A SARS-CoV-2 protease inhibitor was able to ameliorate the infection of kidney cells by SARS-CoV-2. Our results suggest that SARS-CoV-2 can directly infect kidney cells and induce cell injury with subsequent fibrosis. These data could explain both acute kidney injury in COVID-19 patients and the development of chronic kidney disease in long COVID.
Collapse
|
25
|
Mao Z, Tan Y, Tao J, Li L, Wang H, Yu F, Perl A, Zhao M. Renal mTORC1 activation is associated with disease activity and prognosis in lupus nephritis. Rheumatology (Oxford) 2022; 61:3830-3840. [PMID: 35040950 DOI: 10.1093/rheumatology/keac037] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/11/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE This study was initiated to evaluate mammalian target of rapamycin (mTOR) activation in renal tissue of lupus nephritis (LN) patients. METHODS This retrospective study included 187 LN patients, 20 diabetic nephropathy (DN) patients, 10 minimal change disease (MCD) patients, and 10 normal controls (NCs). 7 of 187 LN patients had repeated renal biopsies. mTORC1/2 activation was evaluated by immunohistochemistry and multiplexed immunofluorescence. The association of mTORC1/2 activation with the clinicopathologic indices and prognostic outcomes was analysed among 187 LN patients. Proteomics was performed in renal biopsies of 20 LN patients. Proteomics was employed to comprehensively evaluate the impact of mTOR activation on intrarenal gene expression. RESULTS mTORC1/2 was significantly activated in podocytes, mesangial cells, endothelial cells and tubular epithelial cells of LN patients as compared with those with MCD or NC. The glomerular mTORC1 activation was higher in LN patients compared with DN patients. mTORC1, but not mTORC2, activation strongly correlated with serum albumin, complement C3, proteinuria, and the following pathological biomarkers of LN: crescent formation, interstitial inflammation and fibrosis. Moreover, mTORC1 activation was identified as a prognostic marker in LN patients. Bioinformatic analyses of proteomics and immunohistochemical data unveiled increased complement activation, antigen presentation, and phagocytosis in LN patients with mTORC1 activation. CONCLUSION Renal mTORC1 activation could be a biomarker to reveal disease activity and predict clinical prognosis in LN patients.
Collapse
Affiliation(s)
- Zhaomin Mao
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100034, PR. China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China
| | - Juan Tao
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China
| | - Linlin Li
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China
| | - Hui Wang
- Laboratory of Electron Microscopy, Peking University First Hospital, Beijing, PR China
| | - Feng Yu
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China.,Department of Nephrology, Peking University International Hospital, Beijing, 102206, PR. China
| | - Andras Perl
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, New York, Syracuse, NY, 13210, USA
| | - Minghui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital; Peking University Institute of Nephrology; Key laboratory of Renal Disease, Ministry of Health of China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, 100034, PR. China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100034, PR. China
| |
Collapse
|
26
|
Grahammer F, Huber TB, Artunc F. Role of mTOR Signaling for Tubular Function and Disease. Physiology (Bethesda) 2021; 36:350-358. [PMID: 34514872 DOI: 10.1152/physiol.00021.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) forms two distinct intracellular multiprotein complexes that control a multitude of intracellular processes linked to metabolism, proliferation, actin cytoskeleton, and survival. Recent studies have identified the importance of these complexes for transport regulation of ions and nutrients along the entire nephron. First reports could link altered activity of these complexes to certain disease entities, i.e. diabetic nephropathy, acute kidney injury or hyperkalemia.
Collapse
Affiliation(s)
- Florian Grahammer
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ferruh Artunc
- Department of Internal Medicine, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany.,Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University Tübingen, Tübingen, Germany.,German Center for Diabetes Research, University Tübingen, Tübingen, Germany
| |
Collapse
|
27
|
Schlingmann KP, Jouret F, Shen K, Nigam A, Arjona FJ, Dafinger C, Houillier P, Jones DP, Kleinerüschkamp F, Oh J, Godefroid N, Eltan M, Güran T, Burtey S, Parotte MC, König J, Braun A, Bos C, Ibars Serra M, Rehmann H, Zwartkruis FJ, Renkema KY, Klingel K, Schulze-Bahr E, Schermer B, Bergmann C, Altmüller J, Thiele H, Beck BB, Dahan K, Sabatini D, Liebau MC, Vargas-Poussou R, Knoers NV, Konrad M, de Baaij JH. mTOR-Activating Mutations in RRAGD Are Causative for Kidney Tubulopathy and Cardiomyopathy. J Am Soc Nephrol 2021; 32:2885-2899. [PMID: 34607910 PMCID: PMC8806087 DOI: 10.1681/asn.2021030333] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/07/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Over the last decade, advances in genetic techniques have resulted in the identification of rare hereditary disorders of renal magnesium and salt handling. Nevertheless, approximately 20% of all patients with tubulopathy lack a genetic diagnosis. METHODS We performed whole-exome and -genome sequencing of a patient cohort with a novel, inherited, salt-losing tubulopathy; hypomagnesemia; and dilated cardiomyopathy. We also conducted subsequent in vitro functional analyses of identified variants of RRAGD, a gene that encodes a small Rag guanosine triphosphatase (GTPase). RESULTS In eight children from unrelated families with a tubulopathy characterized by hypomagnesemia, hypokalemia, salt wasting, and nephrocalcinosis, we identified heterozygous missense variants in RRAGD that mostly occurred de novo. Six of these patients also had dilated cardiomyopathy and three underwent heart transplantation. We identified a heterozygous variant in RRAGD that segregated with the phenotype in eight members of a large family with similar kidney manifestations. The GTPase RagD, encoded by RRAGD, plays a role in mediating amino acid signaling to the mechanistic target of rapamycin complex 1 (mTORC1). RagD expression along the mammalian nephron included the thick ascending limb and the distal convoluted tubule. The identified RRAGD variants were shown to induce a constitutive activation of mTOR signaling in vitro. CONCLUSIONS Our findings establish a novel disease, which we call autosomal dominant kidney hypomagnesemia (ADKH-RRAGD), that combines an electrolyte-losing tubulopathy and dilated cardiomyopathy. The condition is caused by variants in the RRAGD gene, which encodes Rag GTPase D; these variants lead to an activation of mTOR signaling, suggesting a critical role of Rag GTPase D for renal electrolyte handling and cardiac function.
Collapse
Affiliation(s)
- Karl P. Schlingmann
- Department of General Pediatrics, University Children’s Hospital, Münster, Germany
| | - François Jouret
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital, Liège, Belgium,Interdisciplinary Group of Applied Genoproteomics, Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Kuang Shen
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts,Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts,Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Anukrati Nigam
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Francisco J. Arjona
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Claudia Dafinger
- Department of Pediatrics and Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany,Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - Pascal Houillier
- Cordeliers Research Center, Centre National de la Recherche Scientifique (CNRS), ERL8228, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne University, University of Paris, Paris, France,Department of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), European Hospital Georges Pompidou, Paris, France,Reference Center for Hereditary Renal Diseases in Children and Adults (MARHEA), Paris, France
| | - Deborah P. Jones
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Felix Kleinerüschkamp
- Department of Pediatric Cardiology, University Children’s Hospital, Münster, Germany
| | - Jun Oh
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Godefroid
- Division of Pediatric Nephrology, Saint-Luc University Clinics, Catholic University of Louvain, Brussels, Belgium
| | - Mehmet Eltan
- Department of Pediatric Endocrinology and Diabetes, School of Medicine, Marmara University, Istanbul, Turkey
| | - Tülay Güran
- Department of Pediatric Endocrinology and Diabetes, School of Medicine, Marmara University, Istanbul, Turkey
| | - Stéphane Burtey
- Center for Nephrology and Renal Transplantation, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, Marseille, France
| | - Marie-Christine Parotte
- Division of Nephrology-Dialysis, Department of Internal Medicine, CHR Verviers East Belgium, Verviers, Belgium
| | - Jens König
- Department of General Pediatrics, University Children’s Hospital, Münster, Germany
| | - Alina Braun
- Department of Pediatrics and Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany,Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - Caro Bos
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria Ibars Serra
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Holger Rehmann
- Department of Molecular Cancer Research, Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Fried J.T. Zwartkruis
- Department of Molecular Cancer Research, Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kirsten Y. Renkema
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany,CECAD, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - Carsten Bergmann
- Limbach Genetics, Medizinische Genetik Mainz, Mainz, Germany,Division of Nephrology, Department of Medicine, University Hospital Freiburg, Breisgau, Germany
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Bodo B. Beck
- Institute of Human Genetics, University Hospital Cologne and University of Cologne, Faculty of Medicine, Cologne, Germany,Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine, University Hospital Cologne, Cologne, Germany,Center for Rare Diseases, Medical Faculty, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - Karin Dahan
- Center of Human Genetics, Gosselies, Belgium,Division of Nephrology, Saint-Luc University Clinics, Catholic University of Louvain, Brussels, Belgium
| | - David Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts,Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Max C. Liebau
- Department of Pediatrics and Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany,Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany,Center for Rare Diseases, Medical Faculty, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - Rosa Vargas-Poussou
- Department of Genetics, AP-HP, European Hospital Georges Pompidou, Paris, France
| | - Nine V.A.M. Knoers
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martin Konrad
- Department of General Pediatrics, University Children’s Hospital, Münster, Germany
| | - Jeroen H.F. de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
28
|
Giannaki M, Ludwig C, Heermann S, Roussa E. Regulation of electrogenic Na + /HCO 3 - cotransporter 1 (NBCe1) function and its dependence on m-TOR mediated phosphorylation of Ser 245. J Cell Physiol 2021; 237:1372-1388. [PMID: 34642952 DOI: 10.1002/jcp.30601] [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: 05/07/2021] [Revised: 09/09/2021] [Accepted: 10/01/2021] [Indexed: 11/09/2022]
Abstract
Astrocytes are pivotal responders to alterations of extracellular pH, primarily by regulation of their principal acid-base transporter, the membrane-bound electrogenic Na+ /bicarbonate cotransporter 1 (NBCe1). Here, we describe amammalian target of rapamycin (mTOR)-dependent and NBCe1-mediated astroglial response to extracellular acidosis. Using primary mouse cortical astrocytes, we investigated the effect of long-term extracellular metabolic acidosis on regulation of NBCe1 and elucidated the underlying molecular mechanisms by immunoblotting, biotinylation of surface proteins, intracellular H+ recording using the H+ -sensitive dye 2',7'-bis-(carboxyethyl)-5-(and-6)-carboxyfluorescein, and phosphoproteomic analysis. The results showed significant increase of NBCe1-mediated recovery of intracellular pH from acidification in WT astrocytes, but not in cortical astrocytes from NBCe1-deficient mice. Acidosis-induced upregulation of NBCe1 activity was prevented following inhibition of mTOR signaling by rapamycin. Yet, during acidosis or following exposure of astrocytes to rapamycin, surface protein abundance of NBCe1 remained -unchanged. Mutational analysis in HeLa cells suggested that NBCe1 activity was dependent on phosphorylation state of Ser245 , a residue conserved in all NBCe1 variants. Moreover, phosphorylation state of Ser245 is regulated by mTOR and is inversely correlated with NBCe1 transport activity. Our results identify pSer245 as a novel regulator of NBCe1 functional expression. We propose that context-dependent and mTOR-mediated multisite phosphorylation of serine residues of NBCe1 is likely to be a potent mechanism contributing to the response of astrocytes to acid/base challenges during pathophysiological conditions.
Collapse
Affiliation(s)
- Marina Giannaki
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Medical Faculty, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich (TUM), Freising, Germany
| | - Stephan Heermann
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Medical Faculty, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Eleni Roussa
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Medical Faculty, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| |
Collapse
|
29
|
Jing X, Han J, Zhang J, Chen Y, Yuan J, Wang J, Neo S, Li S, Yu X, Wu J. Long non-coding RNA MEG3 promotes cisplatin-induced nephrotoxicity through regulating AKT/TSC/mTOR-mediated autophagy. Int J Biol Sci 2021; 17:3968-3980. [PMID: 34671212 PMCID: PMC8495387 DOI: 10.7150/ijbs.58910] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
Cis-Diamminedichloroplatinum (II) (DDP)-induced nephrotoxicity (DDPIN) may cause irreversible renal injury associated with high morbidity and mortality. Current standard therapies have not achieved satisfactory clinical outcomes due to unclear molecular and cellular mechanisms. Therefore, exploring potential therapies on DDPIN represents an urgent medical need. Present study characterized the role of lncRNA maternally expressed gene 3 (lnc-MEG3) in the pathogenesis of DDPIN. In both in vitro and in murine models of DDP-induced nephrotoxicity, lnc-MEG3 exacerbated DDPIN by negatively regulating miRNA-126 subsequently causing a decreased AKT/TSC/mTOR-mediated autophagy. By silencing lnc-MEG3 or incorporating miRNA-126 mimetics, the proliferation and migration of DDP-treated cells were restored. In vivo, we identified Paeonol to alleviate DDPIN by the inhibition of lnc-MEG3. Taken together, lnc-MEG3 represents a novel therapeutic target for DDPIN and Paeonol may serve as a promising treatment by inhibiting lnc-MEG3 and its related signaling pathways.
Collapse
Affiliation(s)
- Xu Jing
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, 250000, China
| | - Jinming Han
- Department of Clinical Neuroscience, Karolinska Institutet, S-171 76, Sweden
| | - Junhao Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yi Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Juan Yuan
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 76, Sweden
| | - Jue Wang
- Key Laboratory, The Second Hospital of Shandong University, Jinan, 250000, China
| | - Shiyong Neo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Shuijie Li
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, S-171 76, Sweden
| | - Xueyuan Yu
- Department of Nephrology, Qilu hospital of Shandong University, Jinan, China
| | - Jing Wu
- Department of Pharmacology, The Second Hospital of Shandong University, Jinan, 250000, China
| |
Collapse
|
30
|
High glucose-stimulated enhancer of zeste homolog-2 (EZH2) forces suppression of deptor to cause glomerular mesangial cell pathology. Cell Signal 2021; 86:110072. [PMID: 34224844 DOI: 10.1016/j.cellsig.2021.110072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 11/24/2022]
Abstract
Function of mTORC1 and mTORC2 has emerged as a driver of mesangial cell pathologies in diabetic nephropathy. The mechanism of mTOR activation is poorly understood in this disease. Deptor is a constitutive subunit and a negative regulator of both mTOR complexes. Mechanistic investigation in mesangial cells revealed that high glucose decreased the expression of deptor concomitant with increased mTORC1 and mTORC2 activities, induction of hypertrophy and, expression of fibronectin and PAI-1. shRNAs against deptor mimicked these pathologic outcomes of high glucose. Conversely, overexpression of deptor significantly inhibited all effects of high glucose. To determine the mechanism of deptor suppression, we found that high glucose significantly increased the expression of EZH2, resulting in lysine-27 tri-methylation of histone H3 (H3K27Me3). Employing approaches including pharmacological inhibition, shRNA-mediated downregulation and overexpression of EZH2, we found that EZH2 regulates high glucose-induced deptor suppression along with activation of mTOR, mesangial cell hypertrophy and fibronectin/PAI-1 expression. Moreover, expression of hyperactive mTORC1 reversed shEZH2-mediated inhibition of hypertrophy and expression of fibronectin and PAI-1 by high glucose. Finally, in renal cortex of diabetic mice, we found that enhanced expression of EZH2 is associated with decreased deptor levels and increased mTOR activity and, expression of fibronectin and PAI-1. Together, our findings provide a novel mechanism for mTOR activation via EZH2 to induce mesangial cell hypertrophy and matrix expansion during early progression of diabetic nephropathy. These results suggest a strategy for leveraging the intrinsic effect of deptor to suppress mTOR activity via reducing EZH2 as a novel therapy for diabetic nephropathy.
Collapse
|
31
|
Proximal Tubule mTORC1 Is a Central Player in the Pathophysiology of Diabetic Nephropathy and Its Correction by SGLT2 Inhibitors. Cell Rep 2021; 32:107954. [PMID: 32726619 PMCID: PMC7397516 DOI: 10.1016/j.celrep.2020.107954] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease (DKD) increases the risk for mortality and is the leading cause of end-stage renal disease. Treatment with sodium-glucose cotransporter 2 inhibitors (SGLT2i) attenuates the progression of DKD, especially in patients with advanced kidney disease. Herein, we show that in diabetes, mTORC1 activity is increased in renal proximal tubule cells (RPTCs) along with enhanced tubule-interstitial fibrosis; this is prevented by SGLT2i. Constitutive activation of mTORC1 in RPTCs induces renal fibrosis and failure and abolishes the renal-protective effects of SGLT2i in diabetes. On the contrary, partial inhibition of mTORC1 in RPTCs prevents fibrosis and the decline in renal function. Stimulation of mTORC1 in RPTCs turns on a pro-fibrotic program in the renal cortex, whereas its inhibition in diabetes reverses the alterations in gene expression. We suggest that RPTC mTORC1 is a critical node that mediates kidney dysfunction in diabetes and the protective effects of SGLT2i by regulating fibrogenesis. In diabetes, mTORC1 activity is increased in renal proximal tubule cells (RPTCs) Diabetes and SGLT2i regulate mTORC1 by modulating nutrient transport to RPTCs Inhibition of mTORC1 in RPTCs prevents fibrosis and the decline in renal function RPTC mTORC1 mediates renal fibrosis in diabetes and the beneficial effects of SGLT2i
Collapse
|
32
|
Hasan HF, Rashed LA, El Bakary NM. Concerted outcome of metformin and low dose of radiation in modulation of cisplatin induced uremic encephalopathy via renal and neural preservation. Life Sci 2021; 276:119429. [PMID: 33785333 DOI: 10.1016/j.lfs.2021.119429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
AIM The therapeutic expediency of cisplatin was limited due to its nephrotoxic side effects, so this study planned to assess the nephrotic and neuroprotective impact of metformin (MET) and low-dose radiation (LDR) in cisplatin-prompted kidney injury and uremic encephalopathy (UE). METHODS The effect of the 10-day MET treatment (200 mg/kg, orally) and/or fractionated LDR (0.25 Gy, of the total dose of 0.5 Gy, 1st and 7th day, respectively) on (5 mg/kg, intraperitoneally) cisplatin as a single dose was administered at the 5th day. Serum urea, creatinine and renal kidney injury molecule-1 were measured for the assessment of kidney function. Furthermore, the antioxidant potential in the renal and brain tissues was evaluated through, malondialdehyde and reduced glutathione estimation. Moreover, renal apoptotic markers: AMP-activated protein kinase, lipocalin, B-cell lymphoma 2 associated X protein, B-cell lymphoma 2, P53 and beclin 1 were estimated. UE was evaluated through the determination of serum inflammatory markers: nuclear factor kappa B, tumor-necrosis factor-α and interleukin 1 beta likewise, the cognitive deficits were assessed via forced swimming test, gamma-aminobutyric acid, n-methyl-d-aspartate and neuronal nitric oxide synthases besides AMP-activated protein kinase, light chain 3 and caspase3 levels in rats' cerebella. KEY FINDINGS The obtained results revealed a noticeable improvement in the previously mentioned biochemical factors and behavioral tasks that was reinforced by histopathological examination when using the present remedy. SIGNIFICANCE metformin and low doses of radiation afforded renoprotection and neuroprotection against cisplatin-induced acute uremic encephalopathy.
Collapse
Affiliation(s)
- Hesham Farouk Hasan
- Radiation Biology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Laila A Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nermeen M El Bakary
- Radiation Biology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
33
|
Xiong W, Xiong Z, Song A, Lei C, Ye C, Zhang C. Relieving lipid accumulation through UCP1 suppresses the progression of acute kidney injury by promoting the AMPK/ULK1/autophagy pathway. Am J Cancer Res 2021; 11:4637-4654. [PMID: 33754018 PMCID: PMC7978316 DOI: 10.7150/thno.56082] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Rationale: Acute kidney injury (AKI) is a serious clinical emergency with an acute onset, rapid progression, and poor prognosis. Recent evidence suggests that AKI is accompanied by significant metabolic abnormalities, including alterations in lipid metabolism. However, the specific changes in lipids in AKI, and their role and regulation mechanisms are currently unclear. Methods: Quantitative metabolomics was performed in AKI models to reveal the differences of lipid metabolism-related products. Regulated pathway was detected by western blot, qRT-PCR, immunoblot analysis and immunohistochemistry. Results: The present study systematically analyzes the changes in lipid composition in AKI for the first time and find that the degree of lipid accumulation was highly correlated with uncoupling protein 1 (UCP1). Importantly, relieving lipid accumulation in AKI by upregulating UCP1 can significantly inhibit the progression of AKI through promoting AMPK/ULK1/autophagy pathway. Conclusions: The present findings suggest that lipid accumulation in AKI is directly regulated by UCP1, which can activate cell autophagy and thus significantly inhibit disease progression. It will provide new ideas and targets for the treatment of AKI.
Collapse
|
34
|
Hinden L, Kogot-Levin A, Tam J, Leibowitz G. Pathogenesis of diabesity-induced kidney disease: role of kidney nutrient sensing. FEBS J 2021; 289:901-921. [PMID: 33630415 DOI: 10.1111/febs.15790] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/09/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022]
Abstract
Diabetes kidney disease (DKD) is a major healthcare problem associated with increased risk for developing end-stage kidney disease and high mortality. It is widely accepted that DKD is primarily a glomerular disease. Recent findings however suggest that kidney proximal tubule cells (KPTCs) may play a central role in the pathophysiology of DKD. In diabetes and obesity, KPTCs are exposed to nutrient overload, including glucose, free-fatty acids and amino acids, which dysregulate nutrient and energy sensing by mechanistic target of rapamycin complex 1 and AMP-activated protein kinase, with subsequent induction of tubular injury, inflammation, and fibrosis. Pharmacological treatments that modulate nutrient sensing and signaling in KPTCs, including cannabinoid-1 receptor antagonists and sodium glucose transporter 2 inhibitors, exert robust kidney protective effects. Shedding light on how nutrients are sensed and metabolized in KPTCs and in other kidney domains, and on their effects on signal transduction pathways that mediate kidney injury, is important for understanding the pathophysiology of DKD and for the development of novel therapeutic approaches in DKD and probably also in other forms of kidney disease.
Collapse
Affiliation(s)
- Liad Hinden
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Aviram Kogot-Levin
- Diabetes Unit and Endocrine Service, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Joseph Tam
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Gil Leibowitz
- Diabetes Unit and Endocrine Service, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
35
|
Legaz I, Bernardo MV, Alfaro R, Martínez-Banaclocha H, Galián JA, Jimenez-Coll V, Boix F, Mrowiec A, Salmeron D, Botella C, Parrado A, Moya-Quiles MR, Minguela A, Llorente S, de la Peña-Moral J, Muro M. PCR Array Technology in Biopsy Samples Identifies Up-Regulated mTOR Pathway Genes as Potential Rejection Biomarkers After Kidney Transplantation. Front Med (Lausanne) 2021; 8:547849. [PMID: 33681239 PMCID: PMC7927668 DOI: 10.3389/fmed.2021.547849] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Antibody-mediated rejection (AMR) is the major cause of kidney transplant rejection. The donor-specific human leukocyte antigen (HLA) antibody (DSA) response to a renal allograft is not fully understood yet. mTOR complex has been described in the accommodation or rejection of transplants and integrates responses from a wide variety of signals. The aim of this study was to analyze the expression of the mTOR pathway genes in a large cohort of kidney transplant patients to determine its possible influence on the transplant outcome. Methods: A total of 269 kidney transplant patients monitored for DSA were studied. The patients were divided into two groups, one with recipients that had transplant rejection (+DSA/+AMR) and a second group of recipients without rejection (+DSA/-AMR and -DSA/-AMR, controls). Total RNA was extracted from kidney biopsies and reverse transcribed to cDNA. Human mTOR-PCR array technology was used to determine the expression of 84 mTOR pathway genes. STRING and REVIGO software were used to simulate gene to gene interaction and to assign a molecular function. Results: The studied groups showed a different expression of the mTOR pathway related genes. Recipients that had transplant rejection showed an over-expressed transcript (≥5-fold) of AKT1S1, DDIT4, EIF4E, HRAS, IGF1, INS, IRS1, PIK3CD, PIK3CG, PRKAG3, PRKCB (>12-fold), PRKCG, RPS6KA2, TELO2, ULK1, and VEGFC, compared with patients that did not have rejection. AKT1S1 transcripts were more expressed in +DSA/-AMR biopsies compared with +DSA/+AMR. The main molecular functions of up-regulated gene products were phosphotransferase activity, insulin-like grown factor receptor and ribonucleoside phosphate binding. The group of patients with transplant rejection also showed an under-expressed transcript (≥5-fold) of VEGFA (>15-fold), RPS6, and RHOA compared with the group without rejection. The molecular function of down-regulated gene products such as protein kinase activity and carbohydrate derivative binding proteins was also analyzed. Conclusions: We have found a higher number of over-expressed mTOR pathway genes than under-expressed ones in biopsies from rejected kidney transplants (+DSA/+AMR) with respect to controls. In addition to this, the molecular function of both types of transcripts (over/under expressed) is different. Therefore, further studies are needed to determine if variations in gene expression profiles can act as predictors of graft loss, and a better understanding of the mechanisms of action of the involved proteins would be necessary.
Collapse
Affiliation(s)
- Isabel Legaz
- Department of Legal and Forensic Medicine, Faculty of Medicine, Biomedical Research Institute (IMIB), University of Murcia, Murcia, Spain
| | - María Victoria Bernardo
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Rafael Alfaro
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Helios Martínez-Banaclocha
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Jose Antonio Galián
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Victor Jimenez-Coll
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Francisco Boix
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Anna Mrowiec
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Diego Salmeron
- Departamento de Ciencias Sociosanitarias, Universidad de Murcia, Murcia, Spain
- Centro de Investigación Biomédica en Red (CIBER) Epidemiología y Salud Pública (CIBERESP), Murcia, Spain
- Instituto Murciano de Investigacion Biomédica-Arrixaca, Murcia, Spain
| | - Carmen Botella
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Antonio Parrado
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - María Rosa Moya-Quiles
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Alfredo Minguela
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Santiago Llorente
- Department of Nephrology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Jesús de la Peña-Moral
- Department of Pathology Services, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Manuel Muro
- Department of Immunology, University Clinical Hospital Virgen de la Arrixaca-Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
36
|
Liu W, Yi Y, Zhang C, Zhou B, Liao L, Liu W, Hu J, Xu Q, Chen J, Lu J. The Expression of TRIM6 Activates the mTORC1 Pathway by Regulating the Ubiquitination of TSC1-TSC2 to Promote Renal Fibrosis. Front Cell Dev Biol 2021; 8:616747. [PMID: 33634104 PMCID: PMC7901959 DOI: 10.3389/fcell.2020.616747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/14/2020] [Indexed: 02/04/2023] Open
Abstract
Renal fibrosis is considered as the final pathway of all types of kidney diseases, which can lead to the progressive loss of kidney functions and eventually renal failure. The mechanisms behind are diversified, in which the mammalian target of rapamycin (mTOR) pathway is one of the most important regulatory pathways that accounts for the disease. Several processes that are regulated by the mTOR pathway, such as autophagy, epithelial-mesenchymal transition (EMT), and endoplasmic reticulum (ER) stress, are tightly associated with renal fibrosis. In this study, we have reported that the expression of tripartite motif-containing (TRIM) protein 6, a member of TRIM family protein, was highly expressed in renal fibrosis patients and positively correlated with the severity of renal fibrosis. In our established in vitro and in vivo renal fibrosis models, its expression was upregulated by the Angiotensin II-induced nuclear translocation of nuclear factor-κB (NF-κB) p50 and p65. In HK2 cells, the expression of TRIM6 promoted the ubiquitination of tuberous sclerosis proteins (TSC) 1 and 2, two negative regulators of the mTORC1 pathway. Moreover, the knockdown of TRIM6 was found efficient for alleviating renal fibrosis and inhibiting the downstream processes of EMT and ER in both HK2 cells and 5/6-nephrectomized rats. Clinically, the level of TRIM6, TSC1/2, and NF-κB p50 was found closely related to renal fibrosis. As a result, we have presented the first study on the role of TRIM6 in the mTORC1 pathway in renal fibrosis models and our findings suggested that TRIM6 may be a potential target for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Weiwei Liu
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Yi
- Department of Nephrology, Jing'an District Central Hospital of Shanghai/ Jing'an Branch, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuanfu Zhang
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baojuan Zhou
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin Liao
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenrui Liu
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Hu
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiming Xu
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Chen
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianrao Lu
- Department of Nephrology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
37
|
Gui Y, Dai C. mTOR Signaling in Kidney Diseases. KIDNEY360 2020; 1:1319-1327. [PMID: 35372878 PMCID: PMC8815517 DOI: 10.34067/kid.0003782020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/02/2020] [Indexed: 04/27/2023]
Abstract
The mammalian target of rapamycin (mTOR), a serine/threonine protein kinase, is crucial in regulating cell growth, metabolism, proliferation, and survival. Under physiologic conditions, mTOR signaling maintains podocyte and tubular cell homeostasis. In AKI, activation of mTOR signaling in tubular cells and interstitial fibroblasts promotes renal regeneration and repair. However, constitutive activation of mTOR signaling in kidneys results in the initiation and progression of glomerular hypertrophy, interstitial fibrosis, polycystic kidney disease, and renal cell carcinoma. Here, we summarize the recent studies about mTOR signaling in renal physiology and injury, and discuss the possibility of its use as a therapeutic target for kidney diseases.
Collapse
Affiliation(s)
- Yuan Gui
- Department of Nephrology, University of Connecticut Health Center, Farmington, Connecticut
| | - Chunsun Dai
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
38
|
Jiang M, Bai M, Lei J, Xie Y, Xu S, Jia Z, Zhang A. Mitochondrial dysfunction and the AKI-to-CKD transition. Am J Physiol Renal Physiol 2020; 319:F1105-F1116. [PMID: 33073587 DOI: 10.1152/ajprenal.00285.2020] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Acute kidney injury (AKI) has been widely recognized as an important risk factor for the occurrence and development of chronic kidney disease (CKD). Even milder AKI has adverse consequences and could progress to renal fibrosis, which is the ultimate common pathway for various terminal kidney diseases. Thus, it is urgent to develop a strategy to hinder the transition from AKI to CKD. Some mechanisms of the AKI-to-CKD transition have been revealed, such as nephron loss, cell cycle arrest, persistent inflammation, endothelial injury with vascular rarefaction, and epigenetic changes. Previous studies have elucidated the pivotal role of mitochondria in acute injuries and demonstrated that the fitness of this organelle is a major determinant in both the pathogenesis and recovery of organ function. Recent research has suggested that damage to mitochondrial function in early AKI is a crucial factor leading to tubular injury and persistent renal insufficiency. Dysregulation of mitochondrial homeostasis, alterations in bioenergetics, and organelle stress cross talk contribute to the AKI-to-CKD transition. In this review, we focus on the pathophysiology of mitochondria in renal recovery after AKI and progression to CKD, confirming that targeting mitochondria represents a potentially effective therapeutic strategy for the progression of AKI to CKD.
Collapse
Affiliation(s)
- Mingzhu Jiang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Mi Bai
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Lei
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yifan Xie
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Shuang Xu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
39
|
Tang C, Livingston MJ, Liu Z, Dong Z. Autophagy in kidney homeostasis and disease. Nat Rev Nephrol 2020; 16:489-508. [PMID: 32704047 PMCID: PMC7868042 DOI: 10.1038/s41581-020-0309-2] [Citation(s) in RCA: 321] [Impact Index Per Article: 64.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
Autophagy is a conserved lysosomal pathway for the degradation of cytoplasmic components. Basal autophagy in kidney cells is essential for the maintenance of kidney homeostasis, structure and function. Under stress conditions, autophagy is altered as part of the adaptive response of kidney cells, in a process that is tightly regulated by signalling pathways that can modulate the cellular autophagic flux - mammalian target of rapamycin, AMP-activated protein kinase and sirtuins are key regulators of autophagy. Dysregulated autophagy contributes to the pathogenesis of acute kidney injury, to incomplete kidney repair after acute kidney injury and to chronic kidney disease of varied aetiologies, including diabetic kidney disease, focal segmental glomerulosclerosis and polycystic kidney disease. Autophagy also has a role in kidney ageing. However, questions remain about whether autophagy has a protective or a pathological role in kidney fibrosis, and about the precise mechanisms and signalling pathways underlying the autophagy response in different types of kidney cells and across the spectrum of kidney diseases. Further research is needed to gain insights into the regulation of autophagy in the kidneys and to enable the discovery of pathway-specific and kidney-selective therapies for kidney diseases and anti-ageing strategies.
Collapse
Affiliation(s)
- Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Zhiwen Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Second Xiangya Hospital at Central South University, Changsha, China.
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
40
|
Kumar V, Kurth T, Zheleznova NN, Yang C, Cowley AW. NOX4/H 2O 2/mTORC1 Pathway in Salt-Induced Hypertension and Kidney Injury. Hypertension 2020; 76:133-143. [PMID: 32475313 PMCID: PMC10629473 DOI: 10.1161/hypertensionaha.120.15058] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/27/2020] [Indexed: 11/16/2022]
Abstract
We have reported that a high-salt (4.0% NaCl) dietary intake activates mTORC1 and inhibition of this pathway with rapamycin blunts the chronic phase of salt-induced hypertension and renal injury in Dahl salt-sensitive (SS) rats. In SS rats, high-salt intake is known to increase the renal production of H2O2 by NOX4, the most abundant NOX isoform in the kidney, and the global knockout of NOX4 blunts salt-sensitivity in these rats. Here, we explored the hypothesis that elevations of H2O2 by NOX4 in high-salt fed SS rat stimulate mTORC1 for the full development of salt-induced hypertension and renal injury. Our in vitro studies found that H2O2 activates mTORC1 independent of PI3K/AKT and AMPK pathways. To determine the in vivo relevance of NOX4/H2O2/mTORC1 in the salt-induced hypertension, SS-Nox4 knockout (SSNox4-/-) rats were daily administrated with vehicle/rapamycin fed a high-salt diet for 21 days. Rapamycin treatment of SSNox4-/- rats had shown no augmented effect on the salt-induced hypertension nor upon indices of renal injury. Significant reductions of renal T lymphocyte and macrophage together with inhibition of cell proliferation were observed in rapamycin treated rats suggesting a role of mTORC1 independent of NOX4 in the proliferation of immune cell. Given the direct activation of mTORC1 by H2O2 and absence of any further protection from salt-induced hypertension in rapamycin-treated SSNox4-/- rats, we conclude that NOX4-H2O2 is a major upstream activator of mTORC1 that contributes importantly to salt-induced hypertension and renal injury in the SS rat model.
Collapse
Affiliation(s)
- Vikash Kumar
- From the Department of Physiology, Medical College of Wisconsin, Milwaukee
| | - Theresa Kurth
- From the Department of Physiology, Medical College of Wisconsin, Milwaukee
| | | | - Chun Yang
- From the Department of Physiology, Medical College of Wisconsin, Milwaukee
| | - Allen W Cowley
- From the Department of Physiology, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
41
|
Lu Q, Wang M, Gui Y, Hou Q, Gu M, Liang Y, Xiao B, Zhao AZ, Dai C. Rheb1 protects against cisplatin-induced tubular cell death and acute kidney injury via maintaining mitochondrial homeostasis. Cell Death Dis 2020; 11:364. [PMID: 32404875 PMCID: PMC7221100 DOI: 10.1038/s41419-020-2539-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 01/14/2023]
Abstract
Ras homolog enriched in brain (Rheb1), a small GTPase, plays a crucial role in regulating cell growth, differentiation, and survival. However, the role and mechanisms for Rheb1 in tubular cell survival and acute kidney injury (AKI) remain unexplored. Here we found that Rheb1 signaling was activated in kidney tubule of AKI patients and cisplatin-treated mice. A mouse model of tubule-specific deletion of Rheb1 (Tubule-Rheb1−/−) was generated. Compared to control littermates, Tubule-Rheb1−/− mice were phenotypically normal within 2 months after birth but developed more severe kidney dysfunction, tubular cell death including apoptosis, necroptosis and ferroptosis, mitochondrial defect and less PGC-1α expression after cisplatin injection. In primary cultured tubular cells, Rheb1 ablation exacerbated cisplatin-induced cell death and mitochondrial defect. Furthermore, haploinsufficiency for Tsc1 in tubular cells led to Rheb1 activation and mitigated cisplatin-induced cell death, mitochondrial defect and AKI. Together, this study uncovers that Rheb1 may protect against cisplatin-induced tubular cell death and AKI through maintaining mitochondrial homeostasis.
Collapse
Affiliation(s)
- Qingmiao Lu
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Mingjie Wang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Yuan Gui
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Qing Hou
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Mengru Gu
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Yan Liang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Bo Xiao
- Department of Biology, Southern University of Science and Technology, 518000, Shenzhen, P.R. China
| | - Allan Zijian Zhao
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510515, Guangzhou, P.R. China
| | - Chunsun Dai
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China.
| |
Collapse
|
42
|
Verschuren EHJ, Castenmiller C, Peters DJM, Arjona FJ, Bindels RJM, Hoenderop JGJ. Sensing of tubular flow and renal electrolyte transport. Nat Rev Nephrol 2020; 16:337-351. [DOI: 10.1038/s41581-020-0259-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2020] [Indexed: 02/06/2023]
|
43
|
Podocyte autophagy is associated with foot process effacement and proteinuria in patients with minimal change nephrotic syndrome. PLoS One 2020; 15:e0228337. [PMID: 31978139 PMCID: PMC6980606 DOI: 10.1371/journal.pone.0228337] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a cellular mechanism involved in the bulk degradation of proteins and turnover of organelle. Several studies have shown the significance of autophagy of the renal tubular epithelium in rodent models of tubulointerstitial disorder. However, the role of autophagy in the regulation of human glomerular diseases is largely unknown. The current study aimed to demonstrate morphological evidence of autophagy and its association with the ultrastructural changes of podocytes and clinical data in patients with idiopathic nephrotic syndrome, a disease in which patients exhibit podocyte injury. The study population included 95 patients, including patients with glomerular disease (minimal change nephrotic syndrome [MCNS], n = 41; idiopathic membranous nephropathy [IMN], n = 37) and 17 control subjects who underwent percutaneous renal biopsy. The number of autophagic vacuoles and the grade of foot process effacement (FPE) in podocytes were examined by electron microscopy (EM). The relationships among the expression of autophagic vacuoles, the grade of FPE, and the clinical data were determined. Autophagic vacuoles were mainly detected in podocytes by EM. The microtubule-associated protein 1 light chain 3 (LC3)-positive area was co-localized with the Wilms tumor 1 (WT1)-positive area on immunofluorescence microscopy, which suggested that autophagy occurred in the podocytes of patients with MCNS. The number of autophagic vacuoles in the podocytes was significantly correlated with the podocyte FPE score (r = -0.443, p = 0.004), the amount of proteinuria (r = 0.334, p = 0.033), and the level of serum albumin (r = -0.317, p = 0.043) in patients with MCNS. The FPE score was a significant determinant for autophagy after adjusting for the age in a multiple regression analysis in MCNS patients (p = 0.0456). However, such correlations were not observed in patients with IMN or in control subjects. In conclusion, the results indicated that the autophagy of podocytes is associated with FPE and severe proteinuria in patients with MCNS. The mechanisms underlying the activation of autophagy in association with FPE in podocytes should be further investigated in order to elucidate the pathophysiology of MCNS.
Collapse
|
44
|
Kaushal GP, Chandrashekar K, Juncos LA, Shah SV. Autophagy Function and Regulation in Kidney Disease. Biomolecules 2020; 10:E100. [PMID: 31936109 PMCID: PMC7022273 DOI: 10.3390/biom10010100] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a dynamic process by which intracellular damaged macromolecules and organelles are degraded and recycled for the synthesis of new cellular components. Basal autophagy in the kidney acts as a quality control system and is vital for cellular metabolic and organelle homeostasis. Under pathological conditions, autophagy facilitates cellular adaptation; however, activation of autophagy in response to renal injury may be insufficient to provide protection, especially under dysregulated conditions. Kidney-specific deletion of Atg genes in mice has consistently demonstrated worsened acute kidney injury (AKI) outcomes supporting the notion of a pro-survival role of autophagy. Recent studies have also begun to unfold the role of autophagy in progressive renal disease and subsequent fibrosis. Autophagy also influences tubular cell death in renal injury. In this review, we reported the current understanding of autophagy regulation and its role in the pathogenesis of renal injury. In particular, the classic mammalian target of rapamycin (mTOR)-dependent signaling pathway and other mTOR-independent alternative signaling pathways of autophagy regulation were described. Finally, we summarized the impact of autophagy activation on different forms of cell death, including apoptosis and regulated necrosis, associated with the pathophysiology of renal injury. Understanding the regulatory mechanisms of autophagy would identify important targets for therapeutic approaches.
Collapse
Affiliation(s)
- Gur P. Kaushal
- Renal Section, Central Arkansas Veterans Healthcare System Little Rock, Arkansas and Division of Nephrology, 4300 W 7th St, Little Rock, AR 72205, USA; (L.A.J.); (S.V.S.)
- Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR 72205, USA;
| | - Kiran Chandrashekar
- Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR 72205, USA;
| | - Luis A. Juncos
- Renal Section, Central Arkansas Veterans Healthcare System Little Rock, Arkansas and Division of Nephrology, 4300 W 7th St, Little Rock, AR 72205, USA; (L.A.J.); (S.V.S.)
- Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR 72205, USA;
| | - Sudhir V. Shah
- Renal Section, Central Arkansas Veterans Healthcare System Little Rock, Arkansas and Division of Nephrology, 4300 W 7th St, Little Rock, AR 72205, USA; (L.A.J.); (S.V.S.)
- Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR 72205, USA;
| |
Collapse
|
45
|
Rahmani S, Defferrari MS, Wakarchuk WW, Antonescu CN. Energetic adaptations: Metabolic control of endocytic membrane traffic. Traffic 2019; 20:912-931. [DOI: 10.1111/tra.12705] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/11/2019] [Accepted: 10/13/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Sadia Rahmani
- Department of Chemistry and BiologyRyerson University Toronto Ontario Canada
| | | | - Warren W. Wakarchuk
- Department of Chemistry and BiologyRyerson University Toronto Ontario Canada
- Department of Biological SciencesUniversity of Alberta Edmonton Alberta Canada
| | - Costin N. Antonescu
- Department of Chemistry and BiologyRyerson University Toronto Ontario Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital Toronto Ontario Canada
| |
Collapse
|
46
|
Bao H, Zhang Q, Liu X, Song Y, Li X, Wang Z, Li C, Peng A, Gong R. Lithium targeting of AMPK protects against cisplatin-induced acute kidney injury by enhancing autophagy in renal proximal tubular epithelial cells. FASEB J 2019; 33:14370-14381. [PMID: 31661633 DOI: 10.1096/fj.201901712r] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Autophagy has been demonstrated to be vital for kidney homeostasis and is centrally implicated in the pathogenesis of cisplatin-induced acute kidney injury (AKI). Lithium is a potent autophagy inducer in a number of cell types. However, it remains uncertain whether its autophagic activity is associated with a beneficial effect on renal tubular cells in AKI. This study aimed to examine the effect of lithium on renal autophagy in cisplatin-induced AKI. Mice or renal proximal tubular epithelial cells in culture were exposed to cisplatin-induced acute injury in the presence or absence of lithium treatment. AKI or tubular cell injury was evaluated, and cell signaling associated with autophagy was examined. Lithium pretreatment prominently ameliorated acute renal tubular damage in mice exposed to cisplatin insult, associated with enhanced autophagy in renal tubules, as assessed by measuring microtubule-associated protein 1A/1B-light chain 3 (LC3)BII/I expression and autophagosome formation. Consistently, in cisplatin-injured renal tubular cells in vitro, lithium enhanced autophagic activities, improved cell viability, and attenuated cell death. Mechanistically, lithium triggered AMPK-α phosphorylation and activation, which in turn positively correlated with the induced expression of autophagy-related molecules, like mammalian target of rapamycin and LC3BII/I. AMPK-α activation is likely required for lithium-induced tubular cell autophagy and protection in cisplatin-induced AKI because blockade of AMPK-α phosphorylation by compound C markedly abrogated lithium-induced autophagosome formation and mitigated the protective effect of lithium on AKI. Our findings suggest that lithium represents a promising therapeutic strategy for protecting renal tubular cells against cisplatin-induced AKI by enhancing autophagy via AMPK-α activation.-Bao, H., Zhang, Q., Liu, X., Song, Y., Li, X., Wang, Z., Li, C., Peng, A., Gong, R. Lithium targeting of AMPK protects against cisplatin-induced acute kidney injury by enhancing autophagy in renal proximal tubular epithelial cells.
Collapse
Affiliation(s)
- Hui Bao
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Nephrology and Clinical Metabolomics, Tongji University School of Medicine, Shanghai, China.,Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Qianyun Zhang
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Nephrology and Clinical Metabolomics, Tongji University School of Medicine, Shanghai, China
| | - Xinying Liu
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Nephrology and Clinical Metabolomics, Tongji University School of Medicine, Shanghai, China
| | - Yaxiang Song
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Nephrology and Clinical Metabolomics, Tongji University School of Medicine, Shanghai, China
| | - Xinhua Li
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Nephrology and Clinical Metabolomics, Tongji University School of Medicine, Shanghai, China
| | - Zhen Wang
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Nephrology and Clinical Metabolomics, Tongji University School of Medicine, Shanghai, China.,Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Changbin Li
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Nephrology and Clinical Metabolomics, Tongji University School of Medicine, Shanghai, China.,Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island, USA
| | - Ai Peng
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Center for Nephrology and Clinical Metabolomics, Tongji University School of Medicine, Shanghai, China
| | - Rujun Gong
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island, USA.,Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, Ohio, USA
| |
Collapse
|
47
|
Abstract
The increasing global prevalence of type 2 diabetes mellitus (T2DM) and chronic kidney disease (CKD) has prompted research efforts to tackle the growing epidemic of diabetic kidney disease (DKD; also known as diabetic nephropathy). The limited success of much of this research might in part be due to the fact that not all patients diagnosed with DKD have renal dysfunction as a consequence of their diabetes mellitus. Patients who present with CKD and diabetes mellitus (type 1 or type 2) can have true DKD (wherein CKD is a direct consequence of their diabetes status), nondiabetic kidney disease (NDKD) coincident with diabetes mellitus, or a combination of both DKD and NDKD. Preclinical studies using models that more accurately mimic these three entities might improve the ability of animal models to predict clinical trial outcomes. Moreover, improved insights into the pathomechanisms that are shared by these entities - including sodium-glucose cotransporter 2 (SGLT2) and renin-angiotensin system-driven glomerular hyperfiltration and tubular hyper-reabsorption - as well as those that are unique to individual entities might lead to the identification of new treatment targets. Acknowledging that the clinical entity of CKD plus diabetes mellitus encompasses NDKD as well as DKD could help solve some of the urgent unmet medical needs of patients affected by these conditions.
Collapse
|
48
|
Khakipoor S, Giannaki M, Theparambil SM, Zecha J, Küster B, Heermann S, Deitmer JW, Roussa E. Functional expression of electrogenic sodium bicarbonate cotransporter 1 (NBCe1) in mouse cortical astrocytes is dependent on S255‐257 and regulated by mTOR. Glia 2019; 67:2264-2278. [DOI: 10.1002/glia.23682] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/23/2019] [Accepted: 07/03/2019] [Indexed: 01/14/2023]
Affiliation(s)
- Shokoufeh Khakipoor
- Department of Molecular Embryology, Faculty of Medicine Institute of Anatomy and Cell Biology, Albert‐Ludwigs‐Universität Freiburg Freiburg Germany
| | - Marina Giannaki
- Department of Molecular Embryology, Faculty of Medicine Institute of Anatomy and Cell Biology, Albert‐Ludwigs‐Universität Freiburg Freiburg Germany
| | - Shefeeq M. Theparambil
- Department of General Zoology, FB Biology University of Kaiserslautern Kaiserslautern Germany
| | - Jana Zecha
- Chair of Proteomics and Bioanalytics Technical University of Munich Freising Germany
| | - Bernhard Küster
- Chair of Proteomics and Bioanalytics Technical University of Munich Freising Germany
- Bavarian Biomolecular Mass Spectrometry Center (BayBioMS) Technical University of Munich Freising Germany
| | - Stephan Heermann
- Department of Molecular Embryology, Faculty of Medicine Institute of Anatomy and Cell Biology, Albert‐Ludwigs‐Universität Freiburg Freiburg Germany
| | - Joachim W. Deitmer
- Department of General Zoology, FB Biology University of Kaiserslautern Kaiserslautern Germany
| | - Eleni Roussa
- Department of Molecular Embryology, Faculty of Medicine Institute of Anatomy and Cell Biology, Albert‐Ludwigs‐Universität Freiburg Freiburg Germany
| |
Collapse
|
49
|
Gui Y, Lu Q, Gu M, Wang M, Liang Y, Zhu X, Xue X, Sun X, He W, Yang J, Zhao AZ, Xiao B, Dai C. Fibroblast mTOR/PPARγ/HGF axis protects against tubular cell death and acute kidney injury. Cell Death Differ 2019; 26:2774-2789. [PMID: 31024074 DOI: 10.1038/s41418-019-0336-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 02/05/2023] Open
Abstract
Kidney fibroblasts play a crucial role in dictating tubular cell fate and the outcome of acute kidney injury (AKI). The underlying mechanisms remain to be determined. Here, we found that mTOR signaling was activated in fibroblasts from mouse kidneys with ischemia/reperfusion injury (IRI). Ablation of fibroblast Rheb or Rictor promoted, while ablation of fibroblast Tsc1 protected against tubular cell death and IRI in mice. In tubular cells cultured with conditioned media (CM) from Rheb-/- or Rictor-/- fibroblasts, less hepatocyte growth factor (HGF) receptor c-met signaling activation or staurosporine-induced cell apoptosis was observed. While CM from Tsc1-/- fibroblasts promoted tubular cell c-met signaling activation and inhibited staurosporine-induced cell apoptosis. In kidney fibroblasts, blocking mTOR signaling downregulated the expression of peroxisome proliferator-activated receptor gamma (PPARγ) and HGF. Downregulating fibroblast HGF expression or blocking tubular cell c-met signaling facilitated tubular cell apoptosis. Notably, renal PPARγ and HGF expression was less in mice with fibroblast Rheb or Rictor ablation, but more in mice with fibroblast Tsc1 ablation than their littermate controls, respectively. Together, these data suggest that mTOR signaling activation in kidney fibroblasts protects against tubular cell death and dictates the outcome of AKI through stimulating PPARγ and HGF expression.
Collapse
Affiliation(s)
- Yuan Gui
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Qingmiao Lu
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Mengru Gu
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Mingjie Wang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Yan Liang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Xingwen Zhu
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Xian Xue
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Xiaoli Sun
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Weichun He
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Junwei Yang
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China
| | - Allan Zijian Zhao
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510515, Guangzhou, China
| | - Bo Xiao
- Neuroscience and Metabolism Research, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Chunsun Dai
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, China.
| |
Collapse
|
50
|
Ramesh J, Ronsard L, Gao A, Venugopal B. Autophagy Intertwines with Different Diseases-Recent Strategies for Therapeutic Approaches. Diseases 2019; 7:diseases7010015. [PMID: 30717078 PMCID: PMC6473623 DOI: 10.3390/diseases7010015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a regular and substantial “clear-out process” that occurs within the cell and that gets rid of debris that accumulates in membrane-enclosed vacuoles by using enzyme-rich lysosomes, which are filled with acids that degrade the contents of the vacuoles. This machinery is well-connected with many prevalent diseases, including cancer, HIV, and Parkinson’s disease. Considering that autophagy is well-known for its significant connections with a number of well-known fatal diseases, a thorough knowledge of the current findings in the field is essential in developing therapies to control the progression rate of diseases. Thus, this review summarizes the critical events comprising autophagy in the cellular system and the significance of its key molecules in manifesting this pathway in various diseases for down- or upregulation. We collectively reviewed the role of autophagy in various diseases, mainly neurodegenerative diseases, cancer, inflammatory diseases, and renal disorders. Here, some collective reports on autophagy showed that this process might serve as a dual performer: either protector or contributor to certain diseases. The aim of this review is to help researchers to understand the role of autophagy-regulating genes encoding functional open reading frames (ORFs) and its connection with diseases, which will eventually drive better understanding of both the progression and suppression of different diseases at various stages. This review also focuses on certain novel therapeutic strategies which have been published in the recent years based on targeting autophagy key proteins and its interconnecting signaling cascades.
Collapse
Affiliation(s)
- Janani Ramesh
- Department of Medical Biochemistry, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India.
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Larance Ronsard
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02140, USA.
| | - Anthony Gao
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bhuvarahamurthy Venugopal
- Department of Medical Biochemistry, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India.
| |
Collapse
|