1
|
Wang Z, Yu X, Ma H, Yao S, Li Z, Zhang R, Liang H, Jiao J. Proprotein convertase subtilisin/kexin type 9 contributes to cisplatin-induced acute kidney injury by interacting with cyclase-associated protein 1 to promote megalin lysosomal degradation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119984. [PMID: 40339661 DOI: 10.1016/j.bbamcr.2025.119984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/17/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Cisplatin-induced acute kidney injury (AKI) is associated with a considerable risk of mortality, highlighting the critical need for effective preventive and therapeutic strategies to mitigate its impact on patients' outcomes. Mounting evidence suggests that administration of the proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor evolocumab significantly reduces the risk of AKI, however, the underlying mechanisms remain poorly understood. Megalin is an endocytic receptor that plays a crucial role in tubular cells. In this study, elevated PCSK9 expression, accompanied by decreased megalin expression, was observed in cellular and murine models of cisplatin-induced AKI. Further experiments revealed that PCSK9 overexpression downregulated megalin expression and promoted tubular injury. Additionally, the PCSK9 inhibitor evolocumab inhibited megalin loss and protected against increases in urinary protein levels, blood urea nitrogen, serum creatinine, and the kidney injury markers neutrophil gelatinase-associated lipocalin and kidney injury molecule 1. Mechanistically, PCSK9 binds to megalin and facilitates its lysosomal degradation through the coordinated actions of cyclase-associated protein 1 (CAP1) and human leukocyte antigen C (HLA-C). Similar to evolocumab, CAP1 deletion significantly protected against megalin loss and mitigated tubular injury both in vitro and in vivo. Collectively, these findings suggest that PCSK9 and CAP1 are potential therapeutic targets for patients with cisplatin-induced AKI.
Collapse
Affiliation(s)
- Zihan Wang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, China
| | - Xinying Yu
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, China
| | - Huimin Ma
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, China
| | - Shuang Yao
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, China
| | - Zongda Li
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, China
| | - Rui Zhang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, China
| | - Haihai Liang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, China; College of Pharmacy, Harbin Medical University, 150086 Harbin, China
| | - Jundong Jiao
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, 150086 Harbin, China; Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, 150086 Harbin, China.
| |
Collapse
|
2
|
Hu M, Qin Y, Jiao X. Can ferroptosis be a target for reproductive health? Trends Endocrinol Metab 2025; 36:398-402. [PMID: 39706760 DOI: 10.1016/j.tem.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Ferroptosis has been implicated in several reproductive disorders, but the underlying mechanisms remain unknown; thus, interventions targeting this pathway are lacking. Here we summarize the emerging findings on ferroptosis in reproductive biology and corresponding disorders, and highlight perspectives and challenges on future ferroptosis research with potential clinical applications.
Collapse
Affiliation(s)
- Mengchun Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
| | - Xue Jiao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China.
| |
Collapse
|
3
|
Chen S, Liu R, Mo CK, Wendl MC, Houston A, Lal P, Zhao Y, Caravan W, Shinkle AT, Abedin-Do A, Naser Al Deen N, Sato K, Li X, Targino da Costa ALN, Li Y, Karpova A, Herndon JM, Artyomov MN, Rubin JB, Jain S, Li X, Stewart SA, Ding L, Chen F. Multi-omic and spatial analysis of mouse kidneys highlights sex-specific differences in gene regulation across the lifespan. Nat Genet 2025; 57:1213-1227. [PMID: 40259083 PMCID: PMC12081296 DOI: 10.1038/s41588-025-02161-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 03/11/2025] [Indexed: 04/23/2025]
Abstract
There is a sex bias in the incidence and progression of many kidney diseases. To better understand such sexual dimorphism, we integrated data from six platforms, characterizing 76 kidney samples from 68 mice at six developmental and adult time points, creating a molecular atlas of the mouse kidney across the lifespan for both sexes. We show that proximal tubules have the most sex-biased differentially expressed genes emerging after 3 weeks of age and are associated with hormonal regulations. We reveal potential mechanisms involving both direct and indirect regulation by androgens and estrogens. Spatial profiling identifies distinct sex-biased spatial patterns in the cortex and outer stripe of the outer medulla. Additionally, older mice exhibit more aging-related gene alterations in loops of Henle, proximal tubules and collecting ducts in a sex-dependent manner. Our results enhance the understanding of spatially resolved gene expression and hormone regulation underlying kidney sexual dimorphism across the lifespan.
Collapse
Affiliation(s)
- Siqi Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Ruiyang Liu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Chia-Kuei Mo
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael C Wendl
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrew Houston
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Preet Lal
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Yanyan Zhao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Wagma Caravan
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrew T Shinkle
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Atieh Abedin-Do
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Nataly Naser Al Deen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Kazuhito Sato
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Xiang Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - André Luiz N Targino da Costa
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Alla Karpova
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - John M Herndon
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Joshua B Rubin
- Department of Paediatrics, Washington University School of Medicine St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine St Louis, St. Louis, MO, USA
| | - Sanjay Jain
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Paediatrics, Washington University School of Medicine St Louis, St. Louis, MO, USA
| | - Xue Li
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sheila A Stewart
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA.
| | - Feng Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
4
|
Zhang W, Li R, Lu D, Wang X, Wang Q, Feng X, Qi S, Zhang X. Phospholipids and peroxisomes in ferroptosis: the therapeutic target of acupuncture regulating vascular cognitive impairment and dementia. Front Aging Neurosci 2025; 17:1512980. [PMID: 40365351 PMCID: PMC12070441 DOI: 10.3389/fnagi.2025.1512980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
Ferroptosis, since its conceptualization in 2012, has witnessed an exponential growth in research interest over recent years. It is regulated by various cellular metabolic pathways during chronic cerebral ischemia and hypoxia, including reactive oxygen species (ROS) generation, iron accumulation, abnormalities in glutathione metabolism, and disruptions in lipid and glucose metabolism. With the deepening and widespread research, ferroptosis has emerged as a critical pathway in the pathogenesis of vascular cognitive impairment and dementia (VCID). This unique cell death pathway caused by iron-dependent phospholipid peroxidation is strongly related to VICD. We examine the impact of phospholipid composition on neuronal susceptibility to ferroptosis, with a particular focus on the critical role of polyunsaturated fatty acids (PUFAs) in this process. Intriguingly, peroxisomes, as key regulators of lipid metabolism and oxidative stress, influence the susceptibility of neuronal cells to ferroptosis through the synthesis of plasmalogens and other lipid species. In this Review, we provide a critical analysis of the current molecular mechanisms and regulatory networks of acupuncture for ferroptosis, the potential functions of acupuncture in peroxisomal functions and phospholipid metabolism, and its neuroprotective effects in VCID, together with a potential for therapeutic targeting. As such, this highlights the theoretical basis for the application of acupuncture in VCID through multi-target regulation of ferroptosis. This review underscores the potential of acupuncture as a non-pharmacological therapeutic approach in VCID, offering new insights into its role in modulating ferroptosis and associated metabolic pathways for neuroprotection.
Collapse
Affiliation(s)
- Wenyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruiyu Li
- Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen, China
| | - Donglei Lu
- Sports Training Academy of Tianjin University of Sport, Tianjin, China
| | - Xinliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiuxuan Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuyang Feng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Sai Qi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuezhu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
5
|
Mishima E, Nakamura T, Doll S, Proneth B, Fedorova M, Pratt DA, Friedmann Angeli JP, Dixon SJ, Wahida A, Conrad M. Recommendations for robust and reproducible research on ferroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00843-2. [PMID: 40204928 DOI: 10.1038/s41580-025-00843-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
Ferroptosis is a necrotic, non-apoptotic cell death modality triggered by unrestrained iron-dependent lipid peroxidation. By unveiling the regulatory mechanisms of ferroptosis and its relevance to various diseases, research over the past decade has positioned ferroptosis as a promising therapeutic target. The rapid growth of this research field presents challenges, associated with potentially inadequate experimental approaches that may lead to misinterpretations in the assessment of ferroptosis. Typical examples include assessing whether an observed phenotype is indeed linked to ferroptosis, and selecting appropriate animal models and small-molecule modulators of ferroptotic cell death. This Expert Recommendation outlines state-of-the-art methods and tools to reliably study ferroptosis and increase the reproducibility and robustness of experimental results. We present highly validated compounds and animal models, and discuss their advantages and limitations. Furthermore, we provide an overview of the regulatory mechanisms and the best-studied players in ferroptosis regulation, such as GPX4, FSP1, SLC7A11 and ACSL4, discussing frequent pitfalls in experimental design and relevant guidance. These recommendations are intended for researchers at all levels, including those entering the expanding and exciting field of ferroptosis research.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Sebastian Doll
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
- Translational Redox Biology, TUM Natural School of Sciences, Technical University of Munich, Garching, Germany.
| |
Collapse
|
6
|
Lee WC, Dixon SJ. Mechanisms of ferroptosis sensitization and resistance. Dev Cell 2025; 60:982-993. [PMID: 40199240 DOI: 10.1016/j.devcel.2025.02.004] [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: 09/06/2024] [Revised: 12/08/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis is an iron-dependent and oxidative form of non-apoptotic cell death with roles in development, homeostasis, and disease. Ferroptosis sensitivity can vary between cells, often for reasons that are not well understood. In this perspective, we describe the core ferroptosis mechanism and outline how changes in iron, redox, and lipid metabolism can alter ferroptosis sensitivity. We propose the concept of a ferroptosis sensitivity-resistance continuum to describe how different intrinsic and extrinsic factors interact to push cells toward a more ferroptosis-sensitive or ferroptosis-resistant state, with effects on development and diseases such as cancer.
Collapse
Affiliation(s)
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
Li S, Nordick KV, Elsenousi AE, Bhattacharya R, Kirby RP, Hassan AM, Hochman-Mendez C, Rosengart TK, Liao KK, Mondal NK. Warm-ischemia and cold storage induced modulation of ferroptosis observed in human hearts donated after circulatory death and brain death. Am J Physiol Heart Circ Physiol 2025; 328:H923-H936. [PMID: 40062653 DOI: 10.1152/ajpheart.00806.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/09/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025]
Abstract
We investigated ferroptosis, a type of programmed cell death mechanism, in human hearts donated after brain death (DBD) and those donated after circulatory death (DCD), focusing on warm ischemia time (WIT) and cold storage. A total of 24 hearts were procured, with six from the DBD group and 18 from the DCD group. The DCD group was divided into three subgroups, each containing six hearts, based on different WITs of 20, 40, and 60 min. All procured hearts were placed in cold storage for up to 6 h. Left ventricular biopsies were performed at 0, 2, 4, and 6 h. We measured ferroptosis regulators [glutathione peroxidase 4 (GPX4), acyl-CoA synthetase long chain family member 4 (ACSL4), and transferrin receptor], iron content (Fe2+ and Fe3+), and lipid peroxidation (malondialdehyde, MDA) in the cardiac tissue. Modulation of ferroptosis was observed in both DBD and DCD hearts. Warm ischemia injury increased myocardial vulnerability to ferroptotic cell death. For DBD hearts, up to 6 h of cold storage increases cardiac levels of MDA, iron content, and ACSL4, thereby increasing vulnerability to ferroptotic cell death. In contrast, for DCD hearts with a WIT of 40 min or more, warm ischemia injury was identified as the primary factor contributing to increased myocardial susceptibility to ferroptotic cell death. Ferroptosis may serve as a promising target to optimize cold preservation for DBD hearts. For DCD hearts, strategies to inhibit ferroptosis should focus on the early warm ischemia phase to assess donor heart quality and suitability for transplantation.NEW & NOTEWORTHY The first human heart research explored the effects of ischemia on the myocardial ferroptotic cell death mechanism. Prolonged cold storage increases the susceptibility of DBD hearts to ferroptotic cell death. In contrast, warm ischemic injury appears to be the main factor leading to the vulnerability of DCD heart ferroptosis. Targeting ferroptosis could be beneficial in optimizing cold preservation for DBD hearts. However, for DCD hearts, interventions should focus on the early phase of warm ischemia.
Collapse
Affiliation(s)
- Shiyi Li
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Katherine V Nordick
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Abdussalam E Elsenousi
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Rishav Bhattacharya
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Randall P Kirby
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Adel M Hassan
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Camila Hochman-Mendez
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, Texas, United States
| | - Todd K Rosengart
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Kenneth K Liao
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, Texas, United States
| | - Nandan K Mondal
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, Texas, United States
| |
Collapse
|
8
|
Lu M, Zhan Z, Li D, Chen H, Li A, Hu J, Huang Z, Yi B. Protective role of vitamin D receptor against mitochondrial calcium overload from PM 2.5-Induced injury in renal tubular cells. Redox Biol 2025; 80:103518. [PMID: 39891958 PMCID: PMC11836507 DOI: 10.1016/j.redox.2025.103518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025] Open
Abstract
PURPOSE This research explores the consequences of being exposed to PM2.5 contribute to renal injury while also evaluating the protective role of Vitamin D-VDR signaling in alleviating mitochondrial calcium imbalance and oxidative stress in renal tubular cells. METHODS Animal models of chronic PM2.5 exposure were used to simulate environmental conditions in wild type and VDR-overexpressing mice specific to renal tubules. In parallel, HK-2 cell lines were treated with PM2.5 in vitro. Mitochondrial function, calcium concentration, and oxidative stress markers were assessed. VDR activation, achieved through genetic overexpression and paricalcitol, was induced to examine its effect on mitochondrial calcium uniporter (MCU) expression and mitochondrial calcium regulation. RESULTS PM2.5 exposure caused significant mitochondrial damage in renal tubular cells, including mitochondrial calcium overload, increased oxidative stress, reduced membrane potential, and diminished ATP production. Elevated MCU expressions were a key contributor to these disruptions. VDR activation effectively reversed these effects by downregulating MCU, restoring mitochondrial calcium balance, reducing oxidative stress, and improving renal function. CONCLUSION This study shows that activating Vitamin D-VDR signaling shields the kidneys from PM2.5-induced damage by reestablishing mitochondrial calcium balance and lowering oxidative stress via inhibition of the MCU. These results unveil a new protective role of VDR in defending against environmental pollutants and suggest that targeting the MCU could offer a potential therapeutic strategy for treating chronic kidney disease linked to pollution exposure.
Collapse
Affiliation(s)
- Mengqiu Lu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, China
| | - Zishun Zhan
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, China; Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dan Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, China
| | - Hengbing Chen
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, China
| | - Aimei Li
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, China
| | - Jing Hu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, China
| | - Zhijun Huang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Furong Laboratory, Changsha, Hunan, China.
| | - Bin Yi
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, China.
| |
Collapse
|
9
|
Golestaneh L, Basalely A, Linkermann A, El-Achkar TM, Kim RS, Neugarten J. Sex, Acute Kidney Injury, and Age: A Prospective Cohort Study. Am J Kidney Dis 2025; 85:329-338.e1. [PMID: 39447957 DOI: 10.1053/j.ajkd.2024.10.003] [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: 05/13/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
RATIONALE & OBJECTIVE Animal models of kidney disease suggest a protective role for female sex hormones, but some authorities assert that female sex in humans is a risk factor for acute kidney injury (AKI). To better understand the risk of AKI, we studied the strength of association between sex and AKI incidence in hormonally distinct age groups across the life span. STUDY DESIGN Prospective cohort study. SETTING & PARTICIPANTS All patients hospitalized in the Montefiore Health System between October 15, 2015, and January 1, 2019, excluding those with kidney failure or obstetrics diagnoses. EXPOSURE Male versus female sex. OUTCOME AKI occurring during hospitalization based on KDIGO definitions. ANALYTICAL APPROACH Generalized estimating equation logistic regression adjusted for comorbidities, sociodemographic factors, and severity of illness. Analyses were stratified into 3 age categories: 6 months to≤16 years,>16 years to<55 years, and≥55 years. RESULTS A total of 132,667 individuals were hospitalized a total of 235,629 times. The mean age was 55.2±23.8 (SD) years. The count of hospitalizations for women was 129,912 (55%). Hospitalization count among Black and Hispanic patients was 71,834 (30.5%) and 24,199 (10.3%), respectively. AKI occurred in 53,926 (22.9%) hospitalizations. In adjusted models, there was a significant interaction between age and sex (P<0.001). Boys and men had a higher risk of AKI across all age groups, an association more pronounced in the age group>16 years to<55 years in which the odds ratio for men was 1.7 (95% CI, 1.6-1.8). This age-based pattern remained consistent across prespecified types of hospitalizations. In a sensitivity analysis, women older than 55 years who received prescriptions for estrogen had lower odds of AKI than those without prescriptions. LIMITATIONS Residual confounding. CONCLUSIONS The greatest relative risk of AKI for males occurred during ages>16 to<55 years. The lower risk among postmenopausal women receiving supplemental estrogen supports a protective role for female sex hormones. PLAIN-LANGUAGE SUMMARY Male sex is a risk factor for acute kidney injury (AKI) in animals, but in human studies this association is not as robust. We studied hospitalizations at a single center to examine the association of hospital-acquired AKI and sex. After controlling for various sources of potential bias and stratifying by age categories through the life course, we observed that men have a higher risk of AKI throughout life. This risk was especially high compared with women of fertile age and older women prescribed estrogen. This pattern was consistent in prespecified subgroups of hospitalizations. These results support a protective role for female sex hormones in the occurrence of hospitalized AKI.
Collapse
Affiliation(s)
- Ladan Golestaneh
- Section of Nephrology, Department of Medicine, School of Medicine, Yale University, New Haven, Connecticut; Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York.
| | - Abby Basalely
- Division of Pediatric Nephrology, Department of Pediatrics, Northwell Health, New Hyde Park, Albert Einstein College of Medicine, Bronx, New York
| | - Andreas Linkermann
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York; Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Tarek M El-Achkar
- Division of Nephrology, Department of Medicine, School of Medicine, Indiana University, and the Roudebush Indianapolis VA, Indianapolis, Indiana
| | - Ryung S Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Joel Neugarten
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
10
|
Moronge D, Godley H, Ayulo V, Mellott E, Elgazzaz M, Cooper G, Mohamed R, Ogbi S, Gillis E, Faulkner JL, Sullivan JC. Persistent subclinical renal injury in female rats following renal ischemia-reperfusion injury. Clin Sci (Lond) 2025; 139:CS20241851. [PMID: 39902555 DOI: 10.1042/cs20241851] [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: 08/27/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/05/2025]
Abstract
The incidence of acute kidney injury (AKI) continues to rise in both men and women. Although creatinine levels return to normal quicker in females following AKI than in males, it remains unclear whether subclinical renal injury persists in young females post-AKI. This study tested the hypothesis that AKI results in subclinical renal injury in females despite plasma creatinine returning to sham levels. For the present study, 12-13-week-old female Sprague-Dawley (SD) rats were randomized to sham or 45-minute warm bilateral ischemia-reperfusion surgery as an experimental model of ischemic AKI. Rats were euthanized 1, 3, 7, 14, or 30 days post-AKI/sham. Plasma creatinine, cystatin C, kidney injury molecule 1 (KIM-1), and NGAL were quantified via assay kits or immunoblotting. Kidneys were processed for histological analysis to assess tubular injury and fibrosis, and for electron microscopy to examine mitochondrial morphology. Immunoblots on kidney homogenates were performed to determine oxidative stress and apoptosis. Plasma creatinine levels were increased 24 hours post-AKI but returned to sham control levels three days post-AKI. However, cystatin C, KIM-1, and NGAL were increased 30 days post-AKI compared with sham. Tubular injury, tubulointerstitial fibrosis, and mitochondrial dysfunction were all increased in 30-day post-AKI rats compared with sham. Additionally, 30-day post-AKI rats had higher p-JNK expression and lower antioxidant enzyme glutathione peroxidase and catalase levels compared with sham. AKI resulted in higher expression of cleaved caspase 3, TUNEL+ cells, and caspase 9 than sham. Despite the normalization of creatinine levels, our data support the hypothesis that subclinical renal injury persists following ischemia-reperfusion injury in young female rats.
Collapse
Affiliation(s)
- Desmond Moronge
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Hannah Godley
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Victor Ayulo
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Elisabeth Mellott
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Mona Elgazzaz
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Gibson Cooper
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Riyaz Mohamed
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Safia Ogbi
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Ellen Gillis
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
- Department of Obstetrics & Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, U.S.A
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| |
Collapse
|
11
|
She Y, Guo X, Tan Y, Liu Q, Zhu L, Zhou X, Yu J, Yan Q. Associations of Systemic Immune-Inflammation Index With Mortality Risk Among Adults in Diabetic Kidney Disease, NHANES 1999 to 2018. Can J Diabetes 2025:S1499-2671(25)00019-X. [PMID: 39954995 DOI: 10.1016/j.jcjd.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVES Immune-inflammation plays a crucial role in the pathogenesis of diabetic kidney disease (DKD), but an exact assessment of indicators remains undefined. In this study we address the link between systemic immune-inflammation index (SII) and mortality risk in DKD, and we explore the effect of sex disparities. METHODS Data from patients with DKD from the National Health and Nutritional Examination Surveys (NHANES, 1999 to 2018) were studied and their causes of death were identified from NHANES-related files. A weighted Cox model was used to evaluate hazard ratios for all-cause, cardiovascular, and cardiocerebrovascular mortality, and these associations were visualized by smoothing curves. RESULTS The average SII was 634.20 (103/μL). There were 1,283 deaths recorded during 273,422 person months (396 were cardiovascular related and 461 were cardiocerebrovascular related). Higher SIIs in the fifth quintile were significantly associated with increased mortality (p<0.01). SII trends showed an increased risk of all-cause mortality of >697.0 (103/μL), cardiovascular risk of >717.8 (103/μL), and cardiocerebrovascular risk of >650.0 (103/μL). In men, mortality increased when SII reached 500 to 660 (103/μL) and 700 to 760 (103/μL) for women. CONCLUSIONS There was a significant association between higher SII and increased risk of all-cause, cardiovascular, and cardiocerebrovascular mortality in DKD patients. In addition, although men had lower SII, their mortality was higher than that of women.
Collapse
Affiliation(s)
- Yun She
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiangyun Guo
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Tan
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Qingqing Liu
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingling Zhu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiqiao Zhou
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jiangyi Yu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Qianhua Yan
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
12
|
Husain I, Shah H, Jordan CZ, Natesh NR, Fay OK, Chen Y, Privratsky JR, Kitai H, Souma T, Varghese S, Howell DN, Thorp EB, Luo X. Targeting allograft inflammatory factor 1 reprograms kidney macrophages to enhance repair. J Clin Invest 2025; 135:e185146. [PMID: 39836477 PMCID: PMC11870741 DOI: 10.1172/jci185146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
The role of macrophages (MΦs) remains incompletely understood in kidney injury and repair. The plasticity of MΦs offers an opportunity to polarize them toward mediating injury resolution in both native and transplanted kidneys undergoing ischemia and/or rejection. Here, we show that infiltrating kidney MΦs augmented their own allograft inflammatory factor 1 (AIF-1) expression after injury. Aif1 genetic deletion led to MΦ polarization toward a reparative phenotype while halting the development of kidney fibrosis. The enhanced repair was mediated by higher levels of antiinflammatory and proregenerative markers, leading to a reduction in cell death and an increase in proliferation of kidney tubular epithelial cells after ischemia followed by reperfusion injury (I/RI). Adoptive transfer of Aif1-/- MΦs into Aif1+/+ mice conferred protection against I/RI. Conversely, depletion of MΦs reversed the tissue-reparative effects in Aif1-/- mice. We further demonstrated increased expression of AIF-1 in human kidney biopsies from native kidneys with acute kidney injury or chronic kidney disease, as well as in biopsies from kidney allografts undergoing acute or chronic rejection. We conclude that AIF-1 is a MΦ marker of renal inflammation, and its targeting uncouples MΦ reparative functions from profibrotic functions. Thus, therapies inhibiting AIF-1 when ischemic injury is inevitable have the potential to reduce the global burden of kidney disease.
Collapse
Affiliation(s)
- Irma Husain
- Division of Nephrology, Department of Medicine, and
- Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Holly Shah
- Division of Nephrology, Department of Medicine, and
| | | | - Naveen R. Natesh
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, North Carolina, USA
| | | | | | | | - Hiroki Kitai
- Division of Nephrology, Department of Medicine, and
| | | | - Shyni Varghese
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, North Carolina, USA
- Department of Mechanical Engineering and Materials Science, and
- Department of Orthopaedic Surgery, Duke University, Durham, North Carolina, USA
| | | | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, and
- Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
13
|
Fan Y, Ren Y, Deng L, Lv D, Chen J, Ling Y, Tu J, Xu X, Wang D, Cai Z. Testosterone deficiency aggravates diet-induced non-alcoholic fatty liver disease by inducing hepatocyte ferroptosis via targeting BMAL1 in mice. Int Immunopharmacol 2025; 144:113641. [PMID: 39579542 DOI: 10.1016/j.intimp.2024.113641] [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: 08/23/2024] [Revised: 11/01/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Testosterone deficiency is linked to an increased prevalence of non-alcoholic fatty liver disease (NAFLD), although the mechanisms underlying this association are not fully understood. Ferroptosis, a regulated cell death pathway driven by iron-dependent lipid peroxidation, has been suggested to play a role in NAFLD pathogenesis. Since testosterone deficiency is associated with lipid disorders and iron deposition, we hepothesize that ferroptosis may be involved in the pathogenesis of diet-induced NAFLD exacerbated by testosterone deficiency. METHODS Apolipoprotein E (APOE-/-) mice were subjected to sham surgery or bilateral castration and subsequently fed a high-fat diet for 16 weeks. Liver gene expression was analyzed using RNA sequencing. Additional assessments included blood analysis, histological staining, measurement of iron and antioxidant enzyme levels, quantitative real-time PCR, Western blotting, and electron microscopy. The effects of testosterone on ferroptosis induced by free fatty acids (FFAs) and Erastin were further investigated in HepG2 cells in vitro. RESULTS Testosterone deficiency resulted in increased hepatic lipid accumulation and macrovesicular steatosis in high-fat diet-fed APOE-/- mice, accompanied by hepatic inflammation, fibrosis, and elevated liver enzyme levels. Transcriptomic analysis revealed that testosterone deficiency affects ferroptosis and circadian rhythm-related signaling pathways. Castrated APOE-/- mice exhibited significantly higher hepatic iron deposition, lipid peroxidation, and expression of key ferroptosis-related proteins, along with decreased Brain and muscle ARNT-like gene 1 (BMAL1) protein expression. In vitro, testosterone treatment reduced lipid and iron accumulation and lipid peroxidation in HepG2 cells subjected to FFAs and Erastin. Moreover, BMAL1 knockdown negated the protective effects of testosterone against ferroptosis in hepatocytes. CONCLUSION Our study demonstrated that testosterone deficiency exacerbates NAFLD induced by a high-fat diet by promoting hepatocyte ferroptosis through modulation of the circadian protein BMAL1.
Collapse
Affiliation(s)
- Yingying Fan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yujie Ren
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liqun Deng
- Laboratory Animal Resources Center, Westlake University, Hangzhou, China, 310053
| | - Dongying Lv
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiayan Chen
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yun Ling
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jue Tu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaoping Xu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Dejun Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhaowei Cai
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
14
|
Fuchs MA, Grabner A, Shi M, Murray SL, Burke EJ, Latic N, Thiriveedi V, Roper J, Ide S, Abe K, Kitai H, Souma T, Wolf M. Intestinal Cyp24a1 regulates vitamin D locally independent of systemic regulation by renal Cyp24a1 in mice. J Clin Invest 2024; 135:e179882. [PMID: 39688907 PMCID: PMC11827884 DOI: 10.1172/jci179882] [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: 01/30/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024] Open
Abstract
Vitamin D regulates mineral homeostasis. The most biologically active form of vitamin D, 1,25-dihydroxyvitamin D (1,25D), is synthesized by CYP27B1 from 25-dihydroxyvitamin D (25D) and is inactivated by CYP24A1. Human monogenic diseases and genome-wide association studies support a critical role for CYP24A1 in regulation of mineral homeostasis, but little is known about its tissue-specific effects. Here, we describe the responses of mice with inducible global deletion, kidney-specific, and intestine-specific deletion of Cyp24a1 to dietary calcium challenge and chronic kidney disease (CKD). Global and kidney-specific Cyp24a1 deletion caused similar syndromes of systemic vitamin D intoxication: elevated circulating 1,25D, 25D, and fibroblast growth factor 23 (FGF23), activation of vitamin D target genes in the kidney and intestine, hypercalcemia, and suppressed parathyroid hormone (PTH). In contrast, mice with intestine-specific Cyp24a1 deletion demonstrated activation of vitamin D target genes exclusively in the intestine, despite no changes in systemic vitamin D levels. In response to a high calcium diet, PTH was suppressed, despite normal serum calcium. In mice with CKD, intestinal Cyp24a1 deletion decreased PTH and FGF23 without precipitating hypercalcemia. These results implicate kidney CYP24A1 in systemic vitamin D regulation while independent local effects of intestinal CYP24A1 could be targeted to treat secondary hyperparathyroidism in CKD.
Collapse
Affiliation(s)
- Michaela A.A. Fuchs
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Division of Nephrology, Department of Medicine and
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melody Shi
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Susan L. Murray
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Emily J. Burke
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Nejla Latic
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | | | - Jatin Roper
- Division of Gastroenterology, Department of Medicine and
| | - Shintaro Ide
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Koki Abe
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hiroki Kitai
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tomokazu Souma
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
15
|
Airik M, Clayton K, Wipf P, Airik R. JP4-039 Mitigates Cisplatin-Induced Acute Kidney Injury by Inhibiting Oxidative Stress and Blocking Apoptosis and Ferroptosis in Mice. Antioxidants (Basel) 2024; 13:1534. [PMID: 39765862 PMCID: PMC11727076 DOI: 10.3390/antiox13121534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/15/2025] Open
Abstract
Cisplatin is a commonly used chemotherapeutic agent in the treatment of a wide array of cancers. Due to its active transport into the kidney proximal tubule cells, cisplatin treatment can cause a buildup of this nephrotoxic compound in the kidney, resulting in acute kidney injury (AKI). About 30% of patients receiving cisplatin chemotherapy develop cisplatin-induced AKI. JP4-039 is a mitochondria-targeted reactive oxygen species (ROS) and electron scavenger. Recent studies have shown that JP4-039 mitigates a variety of genotoxic insults in preclinical studies in rodents by suppressing oxidative stress-mediated tissue damage and blocking apoptosis and ferroptosis. However, the benefits of JP4-039 treatment have not been tested in the setting of AKI. In this study, we investigated the potential renoprotective effect of JP4-039 on cisplatin-induced AKI. To address this goal, we treated mice with JP4-039 before or after cisplatin administration and analyzed them for functional and molecular changes in the kidney. JP4-039 co-administration attenuated cisplatin-induced renal dysfunction and histopathological changes. Upregulation of tubular injury markers was also suppressed by JP4-039. Mechanistically, JP4-039 suppressed lipid peroxidation, prevented tissue oxidative stress, and preserved the glutathione levels in cisplatin-injected mice. An increase in cisplatin-induced apoptosis and ferroptosis was also alleviated by the compound. Moreover, JP4-039 inhibited cytokine overproduction in cisplatin-injected mice. Together, our findings demonstrate that JP4-039 is a promising therapeutic agent against cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Merlin Airik
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Kacian Clayton
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Rannar Airik
- Division of Nephrology, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
16
|
Bloomer SA, Wagner BA, Buettner GR, Brown KE. Liver iron stores and effectors of ferroptosis are dependent on age and sex. Exp Physiol 2024; 109:2046-2056. [PMID: 39422319 PMCID: PMC11607622 DOI: 10.1113/ep092035] [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: 05/20/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024]
Abstract
Ferroptosis is a form of cell death characterized by a pro-oxidative cellular milieu and iron-dependent lipid peroxidation. Ferroptosis has been implicated in various forms of liver injury, in keeping with the major role of the liver in iron metabolism. Limited research has addressed potential differences in ferroptosis mediators with age and sex, especially in an in vivo model. The goal of this investigation was to evaluate hepatic labile iron and mediators of ferroptosis with ageing in both sexes. Because female animals generally display greater antioxidant defences than males, we hypothesized that females would display a phenotype resistant to ferroptosis. Here, we determined iron contents, protein expression of ferroptosis mediators and measures of oxidative injury in liver samples from 12- and 24-month-old male and female Fischer 344 rats. In comparison to males, the livers of female rats at both ages contained more non-haem iron, which was associated with greater ferritin heavy chain expression and attenuated expression of transferrin receptor-1. In female rats, the 24-month-old group had higher contents of thiobarbituric acid reactive substances compared with their 12-month-old counterparts, yet similar contents of labile iron. These results suggest a disconnect between labile iron contents and oxidative injury with age. Female animals also displayed greater expression of acyl-CoA synthetase long-chain family member 4 (ACSL4), a modulator of ferroptosis, and greater abundance of high molecular weight 4-hydroxnonenal-modified proteins. These results demonstrate clear differences in iron and ferroptosis mediators between sexes and suggest that female rats of this strain might be more susceptible to ferroptosis.
Collapse
Affiliation(s)
- Steven A. Bloomer
- Division of Science and EngineeringPenn State AbingtonAbingtonPennsylvaniaUSA
| | - Brett A. Wagner
- Free Radical and Radiation Biology, Department of Radiation OncologyUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Garry R. Buettner
- Free Radical and Radiation Biology, Department of Radiation OncologyUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
| | - Kyle E. Brown
- Free Radical and Radiation Biology, Department of Radiation OncologyUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
- Iowa City Veterans Administration Medical CenterIowa CityIowaUSA
- Division of Gastroenterology‐Hepatology, Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
| |
Collapse
|
17
|
Oh SJ, Shin YY, Ahn JS, Park HJ, Kang MJ, Shin TH, Lee BC, Kim WK, Oh JM, Lee D, Kim YH, Kim JM, Sung ES, Lee EW, Jeong JH, Lee BJ, Seo Y, Kim HS. TGFβ2-Driven Ferritin Degradation and Subsequent Ferroptosis Underlie Salivary Gland Dysfunction in Postmenopausal Conditions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400660. [PMID: 39481440 DOI: 10.1002/advs.202400660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/05/2024] [Indexed: 11/02/2024]
Abstract
Despite the high incidence of dry mouth in postmenopausal women, its underlying mechanisms and therapeutic interventions remain underexplored. Using ovariectomized (OVX) mouse models, here this study identifies ferroptosis, an iron-dependent regulated cell death, as a central mechanism driving postmenopausal salivary gland (SG) dysfunction. In the OVX-SGs, TGFβ signaling pathway is enhanced with the aberrant TGFβ2 expression in SG mesenchymal cells. Intriguingly, TGFβ2 treatment reduces iron-storing ferritin levels, leading to lipid peroxidation and ferroptotic death in SG epithelial organoids (SGOs). Mechanistically, TGFβ2 promotes the autophagy-mediated ferritin degradation, so-called ferritinophagy. A notable overexpression of the type III TGFβ receptor (TβRIII) is found in the OVX-SGs and TGFβ2-treated SGOs, while the silencing of TβRIII mitigates the ferroptosis-mediated deleterious effects of TGFβ2 on SGOs. Finally, administration of ferroptosis inhibitor, Liproxstatin-1 (Lip-1), improves saliva secretion in OVX mice. Present findings collectively suggest a link between TGFβ signaling, ferroptosis, and SG injury, offering new therapeutic avenues for postmenopausal xerostomia.
Collapse
Affiliation(s)
- Su-Jeong Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Ye Young Shin
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Seoul, 08590, Republic of Korea
| | - Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hee-Jeong Park
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Tae-Hoon Shin
- Department of Laboratory Animal Medicine, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju-si, 63243, Republic of Korea
| | - Byung-Chul Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Won Kyu Kim
- Natural Product Research Center, Korea Institute of Science andTechnology (KIST), Gangneung, 25451, Republic of Korea
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
- Division of Natural Products Applied Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jung-Min Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Ji Min Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, Republic of Korea
| | - Eui-Suk Sung
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Pusan National University School of Medicine, Yangsan Pusan National University Hospital, Yangsan, 50612, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Byung-Joo Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| |
Collapse
|
18
|
He X, Zhang J, Huang M, Wang J, Yang S, Yu X, Xu Y, Yang W. Serum apolipoprotein H determines ferroptosis resistance by modulating cellular lipid composition. Cell Death Dis 2024; 15:718. [PMID: 39353906 PMCID: PMC11445452 DOI: 10.1038/s41419-024-07099-2] [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: 05/16/2024] [Revised: 09/07/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024]
Abstract
Ferroptosis is a regulated cell death process dependent on iron, triggered by the accumulation of lipid peroxidation. The environmental context significantly impacts cellular sensitivities to ferroptosis. Serum, constituting the extracellular fluid composition in vivo, provides crucial environmental biomolecules. In this study, we investigated the influence of sera on ferroptosis induction, pinpointing the serum protein apolipoprotein H (APOH) as a pivotal inhibitor of ferroptosis. Moreover, we elucidated that APOH suppresses ferroptosis by activating the phosphoinositide 3-kinase (PI3K)-AKT-sterol regulatory element-binding proteins (SREBPs) pathway, thereby elevating stearoyl-CoA desaturase (SCD) levels and augmenting cellular monounsaturated fatty acid-containing phospholipids (MUFA-PLs). Furthermore, ApoHinfer, the peptide derivative of the active region of APOH, mimics its ferroptosis inhibitory activity. Our findings underscore the critical role of serum protein APOH in the inhibition of ferroptosis and indicates potential therapeutic applications in treating cancer and diseases associated with ferroptosis.
Collapse
Affiliation(s)
- Xiang He
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahui Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Masha Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Simin Yang
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
19
|
Su Q, Fu M, Xiao W, Zhao H, Yu Y, Feng J, Liu Y, Wan Y, Yu Y, Zhang C. Sex Differences in Doxorubicin‐Induced Cardiotoxicity: Insights from Transcriptome Analysis. ADVANCED THERAPEUTICS 2024. [DOI: 10.1002/adtp.202400036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Indexed: 01/12/2025]
Abstract
AbstractMale patients have a higher risk of cardiotoxicity following doxorubicin (DOX) treatment than female patients. However, how this difference occurs at the transcriptome level remains unclear, and the mechanisms underlying these differences are understudied. This study aimed to describe the transcriptional patterns of males and females after DOX treatment and explore the possible mechanisms of sexual differences in DOX‐induced cardiotoxicity. Following DOX treatment, male mice exhibit more severe heart damage than female mice. Transcriptome analysis of mice with and without DOX treatment showed that differentially expressed genes (DEGs) are significantly different between males and females. The majority of DEGs are sex‐specific, and more DEGs are identified in males than females. A number of genes, including the oxidation‐related genes Gdf15 and Rbm3, exhibited altered expression either in males or females. Some other genes, including the ferroptosis‐related gene Cd74, changed their expression levels in both sexes, but at different scales. Biochemical experiments suggested that cardiomyocyte oxidation and ferroptosis may contribute to the sexual dimorphism of DOX‐induced cardiotoxicity. In summary, this study shows that, after exposure to DOX, males and females respond differently regarding the expression of hundreds of genes, including Gdf15, Rbm3, and Cd74, possibly explaining the sexual differences in DOX‐induced cardiotoxicity.
Collapse
Affiliation(s)
- Qingqing Su
- Key Laboratory of Medical Electrophysiology Ministry of Education Institute of Cardiovascular Research and Institute of Metabolic Diseases Southwest Medical University Luzhou Sichuan 646000 China
| | - Min Fu
- School of Basic Medical Science Southwest Medical University Luzhou Sichuan 646000 China
| | - Wanli Xiao
- Department of Anesthesiology The Affiliated Hospital Southwest Medical University Luzhou 646000 China
| | - Huan Zhao
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology Ministry of Education Institute of Cardiovascular Research and Institute of Metabolic Diseases Southwest Medical University Luzhou Sichuan 646000 China
| | - Jianguo Feng
- Department of Anesthesiology The Affiliated Hospital Southwest Medical University Luzhou 646000 China
| | - Yulin Liu
- Department of Anesthesiology The Affiliated Hospital Southwest Medical University Luzhou 646000 China
| | - Ying Wan
- School of Basic Medical Science Southwest Medical University Luzhou Sichuan 646000 China
| | - Yajun Yu
- Key Laboratory of Medical Electrophysiology Ministry of Education Institute of Cardiovascular Research and Institute of Metabolic Diseases Southwest Medical University Luzhou Sichuan 646000 China
| | - Chunxiang Zhang
- Key Laboratory of Medical Electrophysiology Ministry of Education Institute of Cardiovascular Research and Institute of Metabolic Diseases Southwest Medical University Luzhou Sichuan 646000 China
- School of Basic Medical Science Southwest Medical University Luzhou Sichuan 646000 China
| |
Collapse
|
20
|
Beamish JA, Watts JA, Dressler GR. Gene regulation in regeneration after acute kidney injury. J Biol Chem 2024; 300:107520. [PMID: 38950862 PMCID: PMC11325799 DOI: 10.1016/j.jbc.2024.107520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/03/2024] Open
Abstract
Acute kidney injury (AKI) is a common condition associated with significant morbidity, mortality, and cost. Injured kidney tissue can regenerate after many forms of AKI. However, there are no treatments in routine clinical practice to encourage recovery. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that orchestrate kidney recovery. The advent of high-throughput sequencing technologies and genetic mouse models has opened an unprecedented window into the transcriptional dynamics that accompany both successful and maladaptive repair. AKI recovery shares similar cell-state transformations with kidney development, which can suggest common mechanisms of gene regulation. Several powerful bioinformatic strategies have been developed to infer the activity of gene regulatory networks by combining multiple forms of sequencing data at single-cell resolution. These studies highlight not only shared stress responses but also key changes in gene regulatory networks controlling metabolism. Furthermore, chromatin immunoprecipitation studies in injured kidneys have revealed dynamic epigenetic modifications at enhancer elements near target genes. This review will highlight how these studies have enhanced our understanding of gene regulation in injury response and regeneration.
Collapse
Affiliation(s)
- Jeffrey A Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason A Watts
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Gregory R Dressler
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
21
|
Wang T, He L, Wang S, Ma D. Association between nonalcoholic steatohepatitis and high serum ferritin levels in type 2 diabetes mellitus. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20231405. [PMID: 39045927 PMCID: PMC11262309 DOI: 10.1590/1806-9282.20231405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVE The aim of this study was to assess the role of elevated serum ferritin levels in the onset, pathological progression and prognosis of nonalcoholic fatty liver disease. Nonalcoholic fatty liver disease has been rapidly increasing worldwide. Despite extensive research on the pathogenesis of nonalcoholic fatty liver disease, a lack of sufficient clinical research on the relationship between nonalcoholic fatty liver disease and serum ferritin levels remains. METHODS We analysed 968 patients with type 2 diabetes mellitus who underwent liver ultrasound examination and had their serum ferritin levels measured. The presence of nonalcoholic fatty liver disease and advanced liver fibrosis was determined through abdominal ultrasound examination and the nonalcoholic fatty liver disease fibrosis score. RESULTS Compared to that in the non-nonalcoholic fatty liver disease group, the presence of hyperferritinemia was significantly more common in the nonalcoholic fatty liver disease group (83.3 vs. 56.3%, p=0.005). When patients with nonalcoholic fatty liver disease were stratified by the nonalcoholic fatty liver disease fibrosis score, those with advanced liver fibrosis exhibited a higher prevalence of hyperferritinemia (56.3, 78.9, and 88.9% for none, simple steatosis, and advanced fibrosis, respectively; p for trend=0.002). In multivariate logistic regression, liver fibrosis was independently associated with hyperferritinemia (odds ratio [OR] 1.45; 95% confidence interval [CI] 1.18-2.02; p=0.014), and this association remained significant in male patients after adjusting for other risk factors (OR 2.66; 95% CI 1.43-5.48; p=0.026). CONCLUSION Identifying nonalcoholic fatty liver disease patients at a risk of developing nonalcoholic steatohepatitis and advanced fibrosis is crucial for implementing timely interventions and improving patient outcomes. This study highlights the potential utility of serum ferritin levels as a serum biomarker for identifying nonalcoholic steatohepatitis patients and those at a risk of late-stage fibrosis, particularly in male patients with nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Tong Wang
- Tianjin Huanghe Hospital, Health Management Center – Tianjin, China
| | - Le He
- Tianjin Chest Hospital, Department of Cardiology – Tianjin, China
| | - Shaoxin Wang
- Tianjin Medical University, Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Department of Geriatrics – Tianjin, China
| | - Dequan Ma
- Tianjin Huanghe Hospital, Health Management Center – Tianjin, China
| |
Collapse
|
22
|
Abstract
Ferroptosis is a non-apoptotic cell death mechanism characterized by iron-dependent membrane lipid peroxidation. Here, we review what is known about the cellular mechanisms mediating the execution and regulation of ferroptosis. We first consider how the accumulation of membrane lipid peroxides leads to the execution of ferroptosis by altering ion transport across the plasma membrane. We then discuss how metabolites and enzymes that are distributed in different compartments and organelles throughout the cell can regulate sensitivity to ferroptosis by impinging upon iron, lipid and redox metabolism. Indeed, metabolic pathways that reside in the mitochondria, endoplasmic reticulum, lipid droplets, peroxisomes and other organelles all contribute to the regulation of ferroptosis sensitivity. We note how the regulation of ferroptosis sensitivity by these different organelles and pathways seems to vary between different cells and death-inducing conditions. We also highlight transcriptional master regulators that integrate the functions of different pathways and organelles to modulate ferroptosis sensitivity globally. Throughout this Review, we highlight open questions and areas in which progress is needed to better understand the cell biology of ferroptosis.
Collapse
Affiliation(s)
- Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA.
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
23
|
Wu L, Xue X, He C, Lai Y, Tong L. Cell death‑related molecules and targets in the progression of urolithiasis (Review). Int J Mol Med 2024; 53:52. [PMID: 38666544 PMCID: PMC11090264 DOI: 10.3892/ijmm.2024.5376] [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: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Urolithiasis is a high‑incidence disease caused by calcium oxalate (mainly), uric acid, calcium phosphate, struvite, apatite, cystine and other stones. The development of kidney stones is closely related to renal tubule cell damage and crystal adhesion and aggregation. Cell death, comprising the core steps of cell damage, can be classified into various types (i.e., apoptosis, ferroptosis, necroptosis and pyroptosis). Different crystal types, concentrations, morphologies and sizes cause tubular cell damage via the regulation of different forms of cell death. Oxidative stress caused by high oxalate or crystal concentrations is considered to be a precursor to a variety of types of cell death. In addition, complex crosstalk exists among numerous signaling pathways and their key molecules in various types of cell death. Urolithiasis is considered a metabolic disorder, and tricarboxylic acid cycle‑related molecules, such as citrate and succinate, are closely related to cell death and the inhibition of stone development. However, a literature review of the associations between kidney stone development, metabolism and various types of cell death is currently lacking, at least to the best of our knowledge. Thus, the present review summarizes the major advances in the understanding of regulated cell death and urolithiasis progression.
Collapse
Affiliation(s)
- Liping Wu
- Department of Pharmacy, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Xiaoyan Xue
- Department of Pharmacy, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Chengwu He
- Department of Urology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, P.R. China
| | - Yongchang Lai
- Department of Urology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, P.R. China
- Department of Pharmaceutical Management, School of Medical Business, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Lingfei Tong
- Department of Pharmacy, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
24
|
Chen X, Tsvetkov AS, Shen HM, Isidoro C, Ktistakis NT, Linkermann A, Koopman WJ, Simon HU, Galluzzi L, Luo S, Xu D, Gu W, Peulen O, Cai Q, Rubinsztein DC, Chi JT, Zhang DD, Li C, Toyokuni S, Liu J, Roh JL, Dai E, Juhasz G, Liu W, Zhang J, Yang M, Liu J, Zhu LQ, Zou W, Piacentini M, Ding WX, Yue Z, Xie Y, Petersen M, Gewirtz DA, Mandell MA, Chu CT, Sinha D, Eftekharpour E, Zhivotovsky B, Besteiro S, Gabrilovich DI, Kim DH, Kagan VE, Bayir H, Chen GC, Ayton S, Lünemann JD, Komatsu M, Krautwald S, Loos B, Baehrecke EH, Wang J, Lane JD, Sadoshima J, Yang WS, Gao M, Münz C, Thumm M, Kampmann M, Yu D, Lipinski MM, Jones JW, Jiang X, Zeh HJ, Kang R, Klionsky DJ, Kroemer G, Tang D. International consensus guidelines for the definition, detection, and interpretation of autophagy-dependent ferroptosis. Autophagy 2024; 20:1213-1246. [PMID: 38442890 PMCID: PMC11210914 DOI: 10.1080/15548627.2024.2319901] [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: 08/25/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 03/07/2024] Open
Abstract
Macroautophagy/autophagy is a complex degradation process with a dual role in cell death that is influenced by the cell types that are involved and the stressors they are exposed to. Ferroptosis is an iron-dependent oxidative form of cell death characterized by unrestricted lipid peroxidation in the context of heterogeneous and plastic mechanisms. Recent studies have shed light on the involvement of specific types of autophagy (e.g. ferritinophagy, lipophagy, and clockophagy) in initiating or executing ferroptotic cell death through the selective degradation of anti-injury proteins or organelles. Conversely, other forms of selective autophagy (e.g. reticulophagy and lysophagy) enhance the cellular defense against ferroptotic damage. Dysregulated autophagy-dependent ferroptosis has implications for a diverse range of pathological conditions. This review aims to present an updated definition of autophagy-dependent ferroptosis, discuss influential substrates and receptors, outline experimental methods, and propose guidelines for interpreting the results.Abbreviation: 3-MA:3-methyladenine; 4HNE: 4-hydroxynonenal; ACD: accidentalcell death; ADF: autophagy-dependentferroptosis; ARE: antioxidant response element; BH2:dihydrobiopterin; BH4: tetrahydrobiopterin; BMDMs: bonemarrow-derived macrophages; CMA: chaperone-mediated autophagy; CQ:chloroquine; DAMPs: danger/damage-associated molecular patterns; EMT,epithelial-mesenchymal transition; EPR: electronparamagnetic resonance; ER, endoplasmic reticulum; FRET: Försterresonance energy transfer; GFP: green fluorescent protein;GSH: glutathione;IF: immunofluorescence; IHC: immunohistochemistry; IOP, intraocularpressure; IRI: ischemia-reperfusion injury; LAA: linoleamide alkyne;MDA: malondialdehyde; PGSK: Phen Green™ SK;RCD: regulatedcell death; PUFAs: polyunsaturated fatty acids; RFP: red fluorescentprotein;ROS: reactive oxygen species; TBA: thiobarbituricacid; TBARS: thiobarbituric acid reactive substances; TEM:transmission electron microscopy.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Andrey S. Tsvetkov
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Han-Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Ciro Isidoro
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | | | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Werner J.H. Koopman
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Shouqing Luo
- Peninsula Medical School, University of Plymouth, Plymouth, UK
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer-University of Liège, Liège, Belgium
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Donna D. Zhang
- Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shinya Toyokuni
- Department of Pathology and Biological Response, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Enyong Dai
- The Second Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Gabor Juhasz
- Biological Research Center, Institute of Genetics, Szeged, Hungary
- Department of Anatomy, Cell and Developmental Biology, Eotvos Lorand University, Budapest, Hungary
| | - Wei Liu
- Department of Orthopedics, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Minghua Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Pediatric Cancer, Changsha, China
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiping Zou
- Departments of Surgery and Pathology, University of Michigan Medical School, Ann Arbor, USA
| | - Mauro Piacentini
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
- National Institute for Infectious Diseases IRCCS “Lazzaro Spallanzani”, Rome, Italy
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yangchun Xie
- Department of Oncology, Central South University, Changsha, Hunan, China
| | - Morten Petersen
- Functional genomics, Department of Biology, Copenhagen University, Denmark
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA, USA
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, USA
| | - Charleen T. Chu
- Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Wilmer Eye lnstitute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer, Villejuif, France; Gustave Roussy Cancer, Villejuif, France
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden, Europe
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Moscow, Russia
| | - Sébastien Besteiro
- LPHI, University Montpellier, CNRS, Montpellier, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | | | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Valerian E. Kagan
- Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York, USA
| | - Guang-Chao Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Scott Ayton
- Florey Institute, University of Melbourne, Parkville, Australia
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University School of Medicine, Bunkyo-ku Tokyo, Japan
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Eric H. Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Medical Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jon D. Lane
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Junichi Sadoshima
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine, Newark, USA
| | - Wan Seok Yang
- Department of Biological Sciences, St. John’s University, New York City, NY, USA
| | - Minghui Gao
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Christian Münz
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Michael Thumm
- Department of Cellular Biochemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Martin Kampmann
- Department of Biochemistry & Biophysics, University of California, San Francisco, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, USA
| | - Di Yu
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, Australia
- Faculty of Medicine, Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - Marta M. Lipinski
- Department of Anesthesiology & Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jace W. Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Herbert J. Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer, Villejuif, France; Gustave Roussy Cancer, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
25
|
Buse M, Cheng M, Jankowski V, Lellig M, Sterzer V, Strieder T, Leuchtle K, Martin IV, Seikrit C, Brinkkoettter P, Crispatzu G, Floege J, Boor P, Speer T, Kramann R, Ostendorf T, Moeller MJ, Costa IG, Stamellou E. Lineage tracing reveals transient phenotypic adaptation of tubular cells during acute kidney injury. iScience 2024; 27:109255. [PMID: 38444605 PMCID: PMC10914483 DOI: 10.1016/j.isci.2024.109255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/05/2023] [Accepted: 02/13/2024] [Indexed: 03/07/2024] Open
Abstract
Tubular injury is the hallmark of acute kidney injury (AKI) with a tremendous impact on patients and health-care systems. During injury, any differentiated proximal tubular cell (PT) may transition into a specific injured phenotype, so-called "scattered tubular cell" (STC)-phenotype. To understand the fate of this specific phenotype, we generated transgenic mice allowing inducible, reversible, and irreversible tagging of these cells in a murine AKI model, the unilateral ischemia-reperfusion injury (IRI). For lineage tracing, we analyzed the kidneys using single-cell profiling during disease development at various time points. Labeled cells, which we defined by established endogenous markers, already appeared 8 h after injury and showed a distinct expression set of genes. We show that STCs re-differentiate back into fully differentiated PTs upon the resolution of the injury. In summary, we show the dynamics of the phenotypic transition of PTs during injury, revealing a reversible transcriptional program as an adaptive response during disease.
Collapse
Affiliation(s)
- Marc Buse
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Mingbo Cheng
- Institute for Computational Genomics, RWTH Aachen University Hospital, Aachen, Germany
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Michaela Lellig
- Institute for Molecular Cardiovascular Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Viktor Sterzer
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Thiago Strieder
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Katja Leuchtle
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Ina V. Martin
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Claudia Seikrit
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Paul Brinkkoettter
- Department II of Internal Medicine and Centre for Molecular Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Giuliano Crispatzu
- Department II of Internal Medicine and Centre for Molecular Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Peter Boor
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Timotheus Speer
- Medical Clinic 4, Nephrology, University of Frankfurt und Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Tammo Ostendorf
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Marcus J. Moeller
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Ivan G. Costa
- Institute for Computational Genomics, RWTH Aachen University Hospital, Aachen, Germany
| | - Eleni Stamellou
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
26
|
Balafa O, Fernandez-Fernandez B, Ortiz A, Dounousi E, Ekart R, Ferro CJ, Mark PB, Valdivielso JM, Del Vecchio L, Mallamaci F. Sex disparities in mortality and cardiovascular outcomes in chronic kidney disease. Clin Kidney J 2024; 17:sfae044. [PMID: 38638550 PMCID: PMC11024840 DOI: 10.1093/ckj/sfae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Indexed: 04/20/2024] Open
Abstract
Sex (biologically determined) and gender (socially constructed) modulate manifestations and prognosis of a vast number of diseases, including cardiovascular disease (CVD) and chronic kidney disease (CKD). CVD remains the leading cause of death in CKD patients. Population-based studies indicate that women present a higher prevalence of CKD and experience less CVD than men in all CKD stages, although this is not as clear in patients on dialysis or transplantation. When compared to the general population of the same sex, CKD has a more negative impact on women on kidney replacement therapy. European women on dialysis or recipients of kidney transplants have life expectancy up to 44.8 and 19.8 years lower, respectively, than their counterparts of similar age in the general population. For men, these figures stand at 37.1 and 16.5 years, representing a 21% to 20% difference, respectively. Hormonal, genetic, societal, and cultural influences may contribute to these sex-based disparities. To gain a more comprehensive understanding of these differences and their implications for patient care, well-designed clinical trials that involve a larger representation of women and focus on sex-related variables are urgently needed. This narrative review emphasizes the importance of acknowledging the epidemiology and prognosis of sex disparities in CVD among CKD patients. Such insights can guide research into the underlying pathophysiological mechanisms, leading to optimized treatment strategies and ultimately, improved clinical outcomes.
Collapse
Affiliation(s)
- Olga Balafa
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | | | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
| | - Evangelia Dounousi
- Nephrology Dept, Faculty of Medicine, University of Ioannina and University Hospital of Ioannina. Ioannina, Greece
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, Faculty of Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Charles J Ferro
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Patrick B Mark
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Jose M Valdivielso
- Vascular and Renal Traslational Research Group, UDETMA, Biomedical Research Institute of Lleida, IRBLleida, Lleida, Spain
| | - Lucia Del Vecchio
- Department of Nephrology and Dialysis, Sant'Anna Hospital, ASST Lariana, Como, Italy
| | - Francesca Mallamaci
- Department of Nephrology, Dialysis, and Transplantation Azienda Ospedaliera ‘Bianchi-Melacrino-Morelli’ & CNR-IFC, Reggio Calabria, Italy
| |
Collapse
|
27
|
Hirata Y, Mishima E. Membrane Dynamics and Cation Handling in Ferroptosis. Physiology (Bethesda) 2024; 39:73-87. [PMID: 38193763 PMCID: PMC11283900 DOI: 10.1152/physiol.00029.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024] Open
Abstract
Ferroptosis, a regulated cell death hallmarked by excessive lipid peroxidation, is implicated in various (patho)physiological contexts. During ferroptosis, lipid peroxidation leads to a diverse change in membrane properties and the dysregulation of ion homeostasis via the cation channels, ultimately resulting in plasma membrane rupture. This review illuminates cellular membrane dynamics and cation handling in ferroptosis regulation.
Collapse
Affiliation(s)
- Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
- Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
28
|
Li S, Han Q, Liu C, Wang Y, Liu F, Pan S, Zuo L, Gao D, Chen K, Feng Q, Liu Z, Liu D. Role of ferroptosis in chronic kidney disease. Cell Commun Signal 2024; 22:113. [PMID: 38347570 PMCID: PMC10860320 DOI: 10.1186/s12964-023-01422-8] [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: 08/14/2023] [Accepted: 12/03/2023] [Indexed: 02/15/2024] Open
Abstract
Chronic kidney disease (CKD) has historically been a significant global health concern, profoundly impacting both life and well-being. In the process of CKD, with the gradual loss of renal function, the incidence of various life-threatening complications, such as cardiovascular diseases, cerebrovascular accident, infection and stroke, is also increasing rapidly. Unfortunately, existing treatments exhibit limited ability to halt the progression of kidney injury in CKD, emphasizing the urgent need to delve into the precise molecular mechanisms governing the occurrence and development of CKD while identifying novel therapeutic targets. Renal fibrosis, a typical pathological feature of CKD, plays a pivotal role in disrupting normal renal structures and the loss of renal function. Ferroptosis is a recently discovered iron-dependent form of cell death characterized by lipid peroxide accumulation. Ferroptosis has emerged as a potential key player in various diseases and the initiation of organ fibrosis. Substantial evidence suggests that ferroptosis may significantly contribute to the intricate interplay between CKD and its progression. This review comprehensively outlines the intricate relationship between CKD and ferroptosis in terms of iron metabolism and lipid peroxidation, and discusses the current landscape of pharmacological research on ferroptosis, shedding light on promising avenues for intervention. It further illustrates recent breakthroughs in ferroptosis-related regulatory mechanisms implicated in the progression of CKD, thereby providing new insights for CKD treatment. Video Abstract.
Collapse
Affiliation(s)
- Shiyang Li
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Qiuxia Han
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Chang Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yixue Wang
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Fengxun Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Shaokang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Dan Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Kai Chen
- Kaifeng Renmin Hospital, Kaifeng, 475000, Henan, People's Republic of China
| | - Qi Feng
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Dongwei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
29
|
Pan H, Sun Y, Qian LH, Liao YN, Gai YZ, Huo YM, Li ZQ, Nie HZ. A Nutrient-Deficient Microenvironment Facilitates Ferroptosis Resistance via the FAM60A-PPAR Axis in Pancreatic Ductal Adenocarcinoma. RESEARCH (WASHINGTON, D.C.) 2024; 7:0300. [PMID: 38314086 PMCID: PMC10836236 DOI: 10.34133/research.0300] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024]
Abstract
Ferroptosis, a nonapoptotic form of cell death, is an emerging potential therapeutic target for various diseases, including cancer. However, the role of ferroptosis in pancreatic cancer remains poorly understood. Pancreatic ductal adenocarcinoma (PDAC) is characterized by a poor prognosis and chemotherapy resistance, attributed to its high Kirsten rats arcomaviral oncogene homolog mutation rate and severe nutritional deficits resulting from a dense stroma. Several studies have linked rat sarcoma (RAS) mutations to ferroptosis, suggesting that inducing ferroptosis may be an effective strategy against oncogenic RAS-bearing tumors. We investigated the role of Family With Sequence Similarity 60 Member A (FAM60A) in this study, a protein closely associated with a poor prognosis and highly expressed in PDAC and tumor tissue from KrasG12D/+;Trp53R172H/+; Pdx1-Cre mice, in regulating ferroptosis, tumor growth, and gemcitabine sensitivity in vitro and in vivo. Our results demonstrate that FAM60A regulates 3 essential metabolic enzymes, ACSL1/4 and GPX4, to protect PDAC cells from ferroptosis. Furthermore, we found that YY1 transcriptionally regulates FAM60A expression by promoting its transcription, and the Hippo-YY1 pathway is restricted in the low-amino-acid milieu in the context of nutrient deprivation, leading to downstream suppression of peroxisome proliferator-activated receptor and ACSL1/4 and activation of GPX4 pathways. Importantly, FAM60A knockdown sensitized PDAC cells to gemcitabine treatment. A new understanding of FAM60A transcriptional regulation pattern in PDAC and its dual function in ferroptosis reliever and chemotherapy resistance is provided by our study. Targeting FAM60A may therefore offer a promising therapeutic approach for PDAC by simultaneously addressing 2 major features of the disease (high RAS mutation rate and tumor microenvironment nutrient deficiency) and preventing tumor cell metabolic adaptation.
Collapse
Affiliation(s)
- Hong Pan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yue Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Li-Heng Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying-Na Liao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan-Zhi Gai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan-Miao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zuo-Qing Li
- Innomodels Biotechnology Co., Ltd., 51 Xinpei Road, Jiading District, Shanghai, China
| | - Hui-Zhen Nie
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
30
|
Beamish JA, Telang AC, McElliott MC, Al-Suraimi A, Chowdhury M, Ference-Salo JT, Otto EA, Menon R, Soofi A, Weinberg JM, Patel SR, Dressler GR. Pax protein depletion in proximal tubules triggers conserved mechanisms of resistance to acute ischemic kidney injury preventing transition to chronic kidney disease. Kidney Int 2024; 105:312-327. [PMID: 37977366 PMCID: PMC10958455 DOI: 10.1016/j.kint.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/18/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Abstract
Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Identifying the molecular and genetic regulators unique to nephron segments that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. Here, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI and found them upregulated after severe AKI and correlated with chronic injury. Surprisingly, proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to pre-conditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of proximal tubule cells in the S3 segment that displayed features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic pre-conditioning, and female sex. Thus, our results identified a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both the injury response and protection from ischemic AKI.
Collapse
Affiliation(s)
- Jeffrey A Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| | - Asha C Telang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Madison C McElliott
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anas Al-Suraimi
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mahboob Chowdhury
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jenna T Ference-Salo
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Edgar A Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Abdul Soofi
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Joel M Weinberg
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sanjeevkumar R Patel
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Gregory R Dressler
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Wang W, Ren X, Chen X, Hong Q, Cai G. Integrin β1-rich extracellular vesicles of kidney recruit Fn1+ macrophages to aggravate ischemia-reperfusion-induced inflammation. JCI Insight 2024; 9:e169885. [PMID: 38258908 PMCID: PMC10906229 DOI: 10.1172/jci.insight.169885] [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: 02/21/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Ischemia-reperfusion injury-induced (IRI-induced) acute kidney injury is accompanied by mononuclear phagocyte (MP) invasion and inflammation. However, systematic analysis of extracellular vesicle-carried (EV-carried) proteins mediating intercellular crosstalk in the IRI microenvironment is still lacking. Multiomics analysis combining single-cell RNA-Seq data of kidney and protein profiling of kidney-EV was used to elucidate the intercellular communication between proximal tubular cells (PTs) and MP. Targeted adhesion and migration of various MPs were caused by the secretion of multiple chemokines as well as integrin β1-rich EV by ischemic-damaged PTs after IRI. These recruited MPs, especially Fn1+ macrophagocyte, amplified the surviving PT's inflammatory response by secreting the inflammatory factors TNF-α, MCP-1, and thrombospondin 1 (THBS-1), which could interact with integrin β1 to promote more MP adhesion and interact with surviving PT to further promote the secretion of IL-1β. However, GW4869 reduced MP infiltration and maintained a moderate inflammatory level likely by blocking EV secretion. Our findings establish the molecular bases by which chemokines and kidney-EV mediate PT-MP crosstalk in early IRI and provide insights into systematic intercellular communication.
Collapse
Affiliation(s)
- Wenjuan Wang
- School of Medicine, Nankai University, Tianjin, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| | - Xuejing Ren
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Henan Key Laboratory of Kidney Disease and Immunology, Zhengzhou, Henan, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| | - Guangyan Cai
- School of Medicine, Nankai University, Tianjin, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| |
Collapse
|
32
|
Dibra D, Gagea M, Qi Y, Chau GP, Su X, Lozano G. p53R245W Mutation Fuels Cancer Initiation and Metastases in NASH-driven Liver Tumorigenesis. CANCER RESEARCH COMMUNICATIONS 2023; 3:2640-2652. [PMID: 38047594 PMCID: PMC10761659 DOI: 10.1158/2767-9764.crc-23-0218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/19/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Obesity is a significant global health concern. Non-alcoholic fatty liver disease and non-alcoholic steatohepatitis (NASH) are common risk factors for hepatocellular carcinoma (HCC) and are closely associated with metabolic comorbidities, including obesity and diabetes. The TP53 tumor suppressor is the most frequently mutated gene in liver cancers, with half of these alterations being missense mutations. These mutations produce highly abundant proteins in cancer cells which have both inhibitory effects on wildtype (WT) p53, and gain-of-function (GOF) activities that contribute to tumor progression. A Western diet increases p53 activity in the liver. To elucidate the functional consequences of Trp53 mutations in a NASH-driven liver tumorigenesis model, we generated somatic mouse models with Trp53 deletion or the missense hotspot mutant p53R245W only in hepatocytes and placed mice on a high-fat, choline-deficient diet. p53R245W in the presence of diet increased fatty liver, compensatory proliferation in the liver parenchyma, and enriched genes of tumor-promoting pathways such as KRAS signaling, MYC, and epithelial-mesenchymal transition when compared with controls in the premalignant liver. Moreover, p53R245W suppressed transcriptional activity of WT p53 in the liver in vivo under metabolic challenges, and shortened survival and doubling of HCC incidence as compared with control heterozygous mice. Complete loss of Trp53 also significantly accelerated liver tumor incidence and lowered time-to-tumor development compared with WT controls. p53R245W GOF properties increased carcinoma initiation, fueled mixed hepatocholangial carcinoma incidence, and tripled metastatic disease. Collectively, our in vivo studies indicate that p53R245W has stronger tumor promoting activities than Trp53 loss in the context of NASH. SIGNIFICANCE Using somatic NASH-driven mouse models with p53 deletion or mutant p53R245W only in hepatocytes, we discovered that p53R245W increased carcinoma initiation, fueled hepatocholangial carcinoma incidence, and tripled metastases.
Collapse
Affiliation(s)
- Denada Dibra
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mihai Gagea
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuan Qi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gilda P. Chau
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
33
|
Wang T, Xiong T, Yang Y, Chen X, Ma Z, Zuo B, Ning D, Zhou B, Song R, Liu X, Wang D. Estradiol-mediated small GTP-binding protein GDP dissociation stimulator induction contributes to sex differences in resilience to ferroptosis in takotsubo syndrome. Redox Biol 2023; 68:102961. [PMID: 38007983 PMCID: PMC10719533 DOI: 10.1016/j.redox.2023.102961] [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: 10/09/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Declining beneficial cardiovascular actions of estradiol (E2) have been associated with disproportionate susceptibility to takotsubo syndrome (TTS) in postmenopausal women. However, the underlying mechanisms between E2 and this marked disproportion remain unclear. SmgGDS (small GTP-binding protein GDP dissociation stimulator), as a key modulator of cardiovascular disease, plays protective roles in reducing oxidative stress and exerts pleiotropic effects of statins. Whether SmgGDS levels are influenced by E2 status and the effect of SmgGDS on sex differences in TTS are poorly understood. METHODS Clinical data were reviewed from TTS inpatients. Echocardiography, immunofluorescence, and immunohistochemistry were performed together with expression analysis to uncover phenotypic and mechanism changes in sex differences in TTS-like wild-type (WT) and SmgGDS± mice. HL-1 cardiomyocytes were used to further examine and validate molecular mechanisms. RESULTS In 14 TTS inpatients, TTS had a higher incidence in postmenopausal women as compared to premenopausal women and men. In murine TTS, female WT mice exhibited higher cardiac SmgGDS levels than male WT mice. Ovariectomy reduced SmgGDS expression in female WT mice similar to that observed in male mice, whereas E2 replacement in these ovariectomized (OVX) female mice reversed this effect. The physiological importance of this sex-specific E2-mediated SmgGDS response is underscored by the disparity in cardiac adaptation to isoproterenol (ISO) stimulation between both sexes of WT mice. E2-mediated SmgGDS induction conferred female protection against TTS-like acute cardiac injury involving ferritinophagy-mediated ferroptosis. No such cardioprotection was observed in male WT mice and OVX female. A causal role for SmgGDS in this sex-specific cardioprotective adaptation was indicated, inasmuch as SmgGDS deficiency abolished E2-modulated cardioprotection against ferritinophagy and aggravates TTS progression in both sexes. Consistently, knockdown of SmgGDS in HL-1 cardiomyocytes exacerbated ferroptosis in a ferritinophagy-dependent manner and abrogated the protective role of E2 against ferritinophagy. Mechanistically, our findings revealed that SmgGDS regulated E2-dependent cardioprotective effects via AMPK/mTOR signaling pathway. SmgGDS deficiency abolished E2-conferred protection against ferritinophagy through activating AMPK/mTOR pathway, while treatment with recombinant SmgGDS in HL-1 cells significantly mitigated this pathway-associated ferritinophagy activity. CONCLUSIONS These results demonstrate that SmgGDS is a central mediator of E2-conferred female cardioprotection against ferritinophagy-mediated ferroptosis in TTS.
Collapse
Affiliation(s)
- Ti Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China; Cardiology Division, Emory University School of Medicine, Atlanta, GA, USA
| | - Ting Xiong
- Division of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuxue Yang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| | - Xiwei Chen
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| | - Ziwei Ma
- Clinical Medical College, Dalian Medical University, Dalian, Liaoning, China
| | - Bangyun Zuo
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| | - Dong Ning
- School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Beibei Zhou
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xuesong Liu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Daxin Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu, China.
| |
Collapse
|
34
|
Xiong L, Liu J, Han SY, Koppitch K, Guo JJ, Rommelfanger M, Miao Z, Gao F, Hallgrimsdottir IB, Pachter L, Kim J, MacLean AL, McMahon AP. Direct androgen receptor control of sexually dimorphic gene expression in the mammalian kidney. Dev Cell 2023; 58:2338-2358.e5. [PMID: 37673062 PMCID: PMC10873092 DOI: 10.1016/j.devcel.2023.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/20/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023]
Abstract
Mammalian organs exhibit distinct physiology, disease susceptibility, and injury responses between the sexes. In the mouse kidney, sexually dimorphic gene activity maps predominantly to proximal tubule (PT) segments. Bulk RNA sequencing (RNA-seq) data demonstrated that sex differences were established from 4 and 8 weeks after birth under gonadal control. Hormone injection studies and genetic removal of androgen and estrogen receptors demonstrated androgen receptor (AR)-mediated regulation of gene activity in PT cells as the regulatory mechanism. Interestingly, caloric restriction feminizes the male kidney. Single-nuclear multiomic analysis identified putative cis-regulatory regions and cooperating factors mediating PT responses to AR activity in the mouse kidney. In the human kidney, a limited set of genes showed conserved sex-linked regulation, whereas analysis of the mouse liver underscored organ-specific differences in the regulation of sexually dimorphic gene expression. These findings raise interesting questions on the evolution, physiological significance, disease, and metabolic linkage of sexually dimorphic gene activity.
Collapse
Affiliation(s)
- Lingyun Xiong
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA; Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Seung Yub Han
- Graduate Program in Genomics and Computational Biology, Biomedical Graduate Studies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kari Koppitch
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Jin-Jin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Megan Rommelfanger
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhen Miao
- Graduate Program in Genomics and Computational Biology, Biomedical Graduate Studies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fan Gao
- Caltech Bioinformatics Resource Center at Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ingileif B Hallgrimsdottir
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Lior Pachter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA 91125, USA
| | - Junhyong Kim
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adam L MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
35
|
Tao H, Dar HY, Tian C, Banerjee S, Glazer ES, Srinivasan S, Zhu L, Pacifici R, He P. Differences in hepatocellular iron metabolism underlie sexual dimorphism in hepatocyte ferroptosis. Redox Biol 2023; 67:102892. [PMID: 37741044 PMCID: PMC10519854 DOI: 10.1016/j.redox.2023.102892] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023] Open
Abstract
Males show higher incidence and severity than females in hepatic injury and many liver diseases, but the mechanisms are not well understood. Ferroptosis, an iron-mediated lipid peroxidation-dependent death, plays an important role in the pathogenesis of liver diseases. We determined whether hepatocyte ferroptosis displays gender difference, accounting for sexual dimorphism in liver diseases. Compared to female hepatocytes, male hepatocytes were much more vulnerable to ferroptosis by iron and pharmacological inducers including RSL3 and iFSP1. Male but not female hepatocytes exhibited significant increases in mitochondrial Fe2+ and mitochondrial ROS (mtROS) contents. Female hepatocytes showed a lower expression of iron importer transferrin receptor 1 (TfR1) and mitochondrial iron importer mitoferrin 1 (Mfrn1), but a higher expression of iron storage protein ferritin heavy chain 1 (FTH1). It is well known that TfR1 expression is positively correlated with ferroptosis. Herein, we showed that silencing FTH1 enhanced while knockdown of Mfrn1 decreased ferroptosis in HepG2 cells. Removing female hormones by ovariectomy (OVX) did not dampen but rather enhanced hepatocyte resistance to ferroptosis. Mechanistically, OVX potentiated the decrease in TfR1 and increase in FTH1 expression. OVX also increased FSP1 expression in ERK-dependent manner. Elevation in FSP1 suppressed mitochondrial Fe2+ accumulation and mtROS production, constituting a novel mechanism of FSP1-mediated inhibition of ferroptosis. In conclusion, differences in hepatocellular iron handling between male and female account, at least in part, for sexual dimorphism in induced ferroptosis of the hepatocytes.
Collapse
Affiliation(s)
- Hui Tao
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Hamid Y Dar
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Cheng Tian
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Somesh Banerjee
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Evan S Glazer
- Departments of Surgery and Cancer Center, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA; Atlanta Veterans Administration Medical Center, Decatur, GA, USA
| | - Liqin Zhu
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA, 30322, USA.
| |
Collapse
|
36
|
Beamish JA, Telang AC, McElliott MC, Al-Suraimi A, Chowdhury M, Ference-Salo JT, Otto EA, Menon R, Soofi A, Weinberg JM, Patel SR, Dressler GR. Pax Protein Depletion in Proximal Tubules Triggers Conserved Mechanisms of Resistance to Acute Ischemic Kidney Injury and Prevents Transition to Chronic Kidney Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.559511. [PMID: 37873377 PMCID: PMC10592940 DOI: 10.1101/2023.10.03.559511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. In this report, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI. We found that Pax2 and Pax8 are upregulated after severe AKI and correlate with chronic injury. Surprisingly, we then discovered that proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to preconditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of S3 proximal tubule cells that display features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic preconditioning, and female sex. Taken together, our results identify a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both injury response and protection from ischemic AKI. TRANSLATIONAL STATEMENT Identifying the molecular and genetic regulators unique to the nephron that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are two homologous nephron-specific transcription factors that are critical for kidney development and physiology. Here we report that proximal-tubule-selective depletion of Pax2 and Pax8 protects against both acute and chronic injury and induces an expression profile in the S3 proximal tubule with common features shared among diverse conditions that protect against ischemia. These findings highlight a new role for Pax proteins as potential therapeutic targets to treat AKI.
Collapse
|
37
|
Liu Y, Zhang X, Cao Y, Chen X, Zhu J, Zou Y. Ferrostatin-1 Ameliorated Oxidative Lipid Damage in LPS-induced Acute Lung Injury. J Surg Res 2023; 290:266-275. [PMID: 37321147 DOI: 10.1016/j.jss.2023.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 04/01/2023] [Accepted: 05/07/2023] [Indexed: 06/17/2023]
Abstract
INTRODUCTION Ferroptosis is a new type of regulated cell death that is characterized by the overwhelming iron-dependent accumulation of lethal lipid reactive oxygen species and is involved in various diseases. However, the relationship between ferroptosis and lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains largely unknown. METHODS In this study, iron metabolism and ferroptosis-related gene mRNA levels in the lung tissues of LPS-induced ALI mice at different time points were detected. Then, the histological, cytokines production, and iron levels of LPS-induced ALI mice with or without the pretreatment of the ferroptosis inhibitor ferrostatin-1 (Fer-1) were measured after mice received the ferroptosis inhibitor ferrostatin-1 (Fer-1) intraperitoneally before LPS administration. Ferroptosis-related protein (GPX4, NRF2, and DPP4) expression was measured in the in vivo and in vitro ALI model. Finally, ROS accumulation and lipid peroxidation was measured in in vivo and in vitro study. RESULTS Our results showed that iron metabolism and ferroptosis-related gene mRNA demonstrated significant variation in LPS-treated pulmonary tissues. The ferroptosis inhibitor Fer-1 markedly attenuated the histologic injuries of the lung tissue and suppressed the production of cytokines in bronchoalveolar lavage fluid (BALF). Fer-1 administration reduced the levels of NRF2 and DPP4 protein induced by the LPS challenge. Furthermore, Fer-1 reversed the tendency of iron metabolism, MDA, SOD, and GSH levels induced by LPS administration in in vivo and in vitro. CONCLUSIONS Taken together, ferroptosis inhibition by ferrostatin-1 alleviated acute lung injury through modulating oxidative lipid damages induced by the LPS challenge.
Collapse
Affiliation(s)
- Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xinyi Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yumeng Cao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xia Chen
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jiali Zhu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Yun Zou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
38
|
Irikura R, Nishizawa H, Nakajima K, Yamanaka M, Chen G, Tanaka K, Onodera M, Matsumoto M, Igarashi K. Ferroptosis model system by the re-expression of BACH1. J Biochem 2023; 174:239-252. [PMID: 37094356 DOI: 10.1093/jb/mvad036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023] Open
Abstract
Ferroptosis is a regulated cell death induced by iron-dependent lipid peroxidation. The heme-responsive transcription factor BTB and CNC homology 1 (BACH1) promotes ferroptosis by repressing the transcription of genes involved in glutathione (GSH) synthesis and intracellular labile iron metabolism, which are key regulatory pathways in ferroptosis. We found that BACH1 re-expression in Bach1-/- immortalized mouse embryonic fibroblasts (iMEFs) can induce ferroptosis upon 2-mercaptoethanol removal, without any ferroptosis inducers. In these iMEFs, GSH synthesis was reduced, and intracellular labile iron levels were increased upon BACH1 re-expression. We used this system to investigate whether the major ferroptosis regulators glutathione peroxidase 4 (Gpx4) and apoptosis-inducing factor mitochondria-associated 2 (Aifm2), the gene for ferroptosis suppressor protein 1, are target genes of BACH1. Neither Gpx4 nor Aifm2 was regulated by BACH1 in the iMEFs. However, we found that BACH1 represses AIFM2 transcription in human pancreatic cancer cells. These results suggest that the ferroptosis regulators targeted by BACH1 may vary across different cell types and animal species. Furthermore, we confirmed that the ferroptosis induced by BACH1 re-expression exhibited a propagating effect. BACH1 re-expression represents a new strategy for inducing ferroptosis after GPX4 or system Xc- suppression and is expected to contribute to future ferroptosis research.
Collapse
Key Words
- BACH1 Abbreviations: AIFM2, apoptosis-inducing factor mitochondria-associated 2; ANOVA, analysis of variance; BACH1, BTB and CNC homology 1; Bach1−/− mice, Bach1 knockout mice; BTB, Broad complex, Tramtrack, Bric-a-brac domain; bZIP, basic leucine zipper; ChIP-seq, chromatin immunoprecipitation sequencing; CNC, Cap‘n’Collar region; DAPI, 4′,6-diamidino-2-phenylindole; DFX, deferasirox; DMSO, dimethyl sulfoxide; EMT, epithelial–mesenchymal transition; Ferr-1, ferrostatin-1; FINs, ferroptosis inducers; FSP1, Ferroptosis suppressor protein 1; Fth1, ferritin heavy chain 1; Ftl, ferritin light chain; GCL, glutamate-cysteine ligase; Gclc, GCL catalytic subunit; Gclm, GCL modifier subunit; GEO, Gene Expression Omnibus; GPX4, glutathione peroxidase 4; GSH, glutathione; HO-1 (Hmox1), heme oxygenase 1; iMEFs, immortalized MEFs; KuO, Kusabira Orange; MAFK, musculoaponeurotic fibrosarcoma oncogene homolog bZIP transcription factor K; mBACH1, Bach1 gene of Mus musculus; 2-ME, 2-mercaptoethanol; MEFs, mouse embryonic fibroblasts; NRF2, nuclear factor-erythroid 2-related factor 2; NSA, necrosulfonamide; PDAC, pancreatic ductal adenocarcinoma; PI, Propidium iodide; Ptgs2, prostaglandin-endoperoxide synthase 2; RSL3, (1S,3R)-RSL3; Slc40a1, solute carrier family 40 member 1; Slc7a11, solute carrier family 7 member 11; TFRC, transferrin receptor 1; Z-VAD.FMK, Benzyloxycarbonyl-Val-Ala-Asp (OMe) fluoromethylketone
- extracellular signal
- ferroptosis
- fibroblasts
- transcription
Collapse
Affiliation(s)
- Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Gladstone Institute of Neurological Disease, Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA
| | - Guan Chen
- Department of Molecular Oncology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Masafumi Onodera
- Gene & Cell Therapy Promotion Center, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
39
|
Yehia A, Sousa RAL, Abulseoud OA. Sex difference in the association between blood alcohol concentration and serum ferritin. Front Psychiatry 2023; 14:1230406. [PMID: 37547205 PMCID: PMC10401063 DOI: 10.3389/fpsyt.2023.1230406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction The sex difference in alcohol use disorder (AUD) is ingrained in distinctive neurobiological responses between men and women, which necessitates further investigation for a more tailored management. Methods Minding the findings of iron dysregulation in AUD and the sex difference in iron homeostasis in multiple physiological and pathological settings, we examined the sex difference in the association between serum ferritin and blood alcohol concentration (BAC) in intoxicated males (n = 125) and females (n = 59). We included patients with both serum ferritin tested of any value and a BAC above the level of detection during the same hospital admission period. We investigated sex difference in the relationship between BAC, serum ferritin and liver enzymes in intoxicated critically ill and noncritically ill patients. Results We found a negative association between serum ferritin and BAC in critically ill, intoxicated females [R2 = 0.44, F(1,14) = 11.02, p = 0.005], with much attenuated serum ferritin in females compared to their male counterparts (194.5 ± 280.4 vs. 806.3 ± 3405.7 ng/L, p = 0.002). We found a positive association between serum ferritin and liver enzymes [alanine transaminase (ALT) and aspartate transferase (AST)] in critically ill intoxicated females [ALT: R2 = 0.48, F(1,10) = 9.1, p = 0.013; AST: R2 = 0.68, F(1,10) = 21.2, p = 0.001] and in noncritically ill intoxicated males [ALT: R2 = 0.1, F(1,83) = 9.4, p = 0.003; AST: R2 = 0.1, F(1,78) = 10.5, p = 0.002]. The effect of BAC on serum ferritin was not mediated by ALT [indirect effect: (B = 0.13, p = 0.1)]. We also found a significant effect of sex, anemia, intensive care unit (ICU) admission and mortality on serum ferritin. Discussion Our results suggest that high BAC in intoxicated female patients is associated with attenuated serum ferritin levels, questioning the role of low serum ferritin in female vulnerability to alcohol.
Collapse
Affiliation(s)
- Asmaa Yehia
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, United States
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ricardo A L Sousa
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Osama A Abulseoud
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, United States
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
| |
Collapse
|
40
|
Guo R, Duan J, Pan S, Cheng F, Qiao Y, Feng Q, Liu D, Liu Z. The Road from AKI to CKD: Molecular Mechanisms and Therapeutic Targets of Ferroptosis. Cell Death Dis 2023; 14:426. [PMID: 37443140 PMCID: PMC10344918 DOI: 10.1038/s41419-023-05969-9] [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: 02/09/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023]
Abstract
Acute kidney injury (AKI) is a prevalent pathological condition that is characterized by a precipitous decline in renal function. In recent years, a growing body of studies have demonstrated that renal maladaptation following AKI results in chronic kidney disease (CKD). Therefore, targeting the transition of AKI to CKD displays excellent therapeutic potential. However, the mechanism of AKI to CKD is mediated by multifactor, and there is still a lack of effective treatments. Ferroptosis, a novel nonapoptotic form of cell death, is believed to have a role in the AKI to CKD progression. In this study, we retrospectively examined the history and characteristics of ferroptosis, summarized ferroptosis's research progress in AKI and CKD, and discussed how ferroptosis participates in regulating the pathological mechanism in the progression of AKI to CKD. Furthermore, we highlighted the limitations of present research and projected the future evolution of ferroptosis. We hope this work will provide clues for further studies of ferroptosis in AKI to CKD and contribute to the study of effective therapeutic targets to prevent the progression of kidney diseases.
Collapse
Affiliation(s)
- Runzhi Guo
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Jiayu Duan
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Shaokang Pan
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Fei Cheng
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Yingjin Qiao
- Blood Purification Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Qi Feng
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| | - Dongwei Liu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| | - Zhangsuo Liu
- Research Institute of Nephrology, Zhengzhou University, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| |
Collapse
|
41
|
Tao H, Dar HY, Tian C, Banerjee S, Glazer ES, Srinivasan S, Zhu L, Pacifici R, He P. Differences in Hepatocellular Iron Metabolism Underlie Sexual Dimorphism in Hepatocyte Ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.24.546395. [PMID: 37425728 PMCID: PMC10327041 DOI: 10.1101/2023.06.24.546395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Males show higher incidence and severity than females in hepatic injury and many liver diseases, but the mechanisms are not well understood. Ferroptosis, an iron-mediated lipid peroxidation-dependent death, plays an important role in the pathogenesis of liver diseases. We determined whether hepatocyte ferroptosis displays gender difference, accounting for sexual dimorphism in liver diseases. Compared to female hepatocytes, male hepatocytes were much more vulnerable to ferroptosis by iron and pharmacological inducers including RSL3 and iFSP1. Male but not female hepatocytes exhibited significant increases in mitochondrial Fe 2+ and mitochondrial ROS (mtROS) contents. Female hepatocytes showed a lower expression of iron importer transferrin receptor 1 (TfR1) and mitochondrial iron importer mitoferrin 1 (Mfrn1), but a higher expression of iron storage protein ferritin heavy chain 1 (FTH1). It is well known that TfR1 expression is positively correlated with ferroptosis. Herein, we showed that silencing FTH1 enhanced while knockdown of Mfrn1 decreased ferroptosis in HepG2 cells. Removing female hormones by ovariectomy (OVX) did not dampen but rather enhanced hepatocyte resistance to ferroptosis. Mechanistically, OVX potentiated the decrease in TfR1 and increase in FTH1 expression. OVX also increased FSP1 expression in ERK-dependent manner. Elevation in FSP1 suppressed mitochondrial Fe 2+ accumulation and mtROS production, constituting a novel mechanism of FSP1-mediated inhibition of ferroptosis. In conclusion, differences in hepatocellular iron handling between male and female account, at least in part, for sexual dimorphism in induced ferroptosis of the hepatocytes.
Collapse
|
42
|
Xiong L, Liu J, Han SY, Koppitch K, Guo JJ, Rommelfanger M, Gao F, Hallgrimsdottir IB, Pachter L, Kim J, MacLean AL, McMahon AP. Direct androgen receptor regulation of sexually dimorphic gene expression in the mammalian kidney. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.06.539585. [PMID: 37205355 PMCID: PMC10187285 DOI: 10.1101/2023.05.06.539585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Mammalian organs exhibit distinct physiology, disease susceptibility and injury responses between the sexes. In the mouse kidney, sexually dimorphic gene activity maps predominantly to proximal tubule (PT) segments. Bulk RNA-seq data demonstrated sex differences were established from 4 and 8 weeks after birth under gonadal control. Hormone injection studies and genetic removal of androgen and estrogen receptors demonstrated androgen receptor (AR) mediated regulation of gene activity in PT cells as the regulatory mechanism. Interestingly, caloric restriction feminizes the male kidney. Single-nuclear multiomic analysis identified putative cis-regulatory regions and cooperating factors mediating PT responses to AR activity in the mouse kidney. In the human kidney, a limited set of genes showed conserved sex-linked regulation while analysis of the mouse liver underscored organ-specific differences in the regulation of sexually dimorphic gene expression. These findings raise interesting questions on the evolution, physiological significance, and disease and metabolic linkage, of sexually dimorphic gene activity.
Collapse
Affiliation(s)
- Lingyun Xiong
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Seung Yub Han
- Graduate Program in Genomics and Computational Biology, Biomedical Graduate Studies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kari Koppitch
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Jin-Jin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Megan Rommelfanger
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Fan Gao
- Caltech Bioinformatics Resource Center at Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Lior Pachter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA 91125, USA
| | - Junhyong Kim
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
- Lead Contact
| |
Collapse
|
43
|
Zhou Z, Li J, Zhang X. Natural Flavonoids and Ferroptosis: Potential Therapeutic Opportunities for Human Diseases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37027486 DOI: 10.1021/acs.jafc.2c08128] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Flavonoids are a class of bioactive phytochemicals containing a core 2-phenylchromone skeleton and are widely found in fruits, vegetables, and herbs. Such natural compounds have gained significant attention due to their various health benefits. Ferroptosis is a recently discovered unique iron-dependent mode of cell death. Unlike traditional regulated cell death (RCD), ferroptosis is associated with excessive lipid peroxidation on cellular membranes. Accumulating evidence suggests that this form of RCD is involved in a variety of physiological and pathological processes. Notably, multiple flavonoids have been shown to be effective in preventing and treating diverse human diseases by regulating ferroptosis. In this review, we introduce the key molecular mechanisms of ferroptosis, including iron metabolism, lipid metabolism, and several major antioxidant systems. Additionally, we summarize the promising flavonoids targeting ferroptosis, which provides novel ideas for the management of diseases such as cancer, acute liver injury, neurodegenerative diseases, and ischemia/reperfusion (I/R) injury.
Collapse
Affiliation(s)
- Zheng Zhou
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Jiye Li
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xiaochuan Zhang
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
44
|
Okazaki Y, Hino K. Iron and Cancer: A Special Issue. Cancers (Basel) 2023; 15:cancers15072097. [PMID: 37046758 PMCID: PMC10093076 DOI: 10.3390/cancers15072097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Iron is an essential element for all organisms, and iron-containing proteins play critical roles in cellular functions [...]
Collapse
|
45
|
Murayama M, Hirata H, Shiraki M, Iovanna JL, Yamaza T, Kukita T, Komori T, Moriishi T, Ueno M, Morimoto T, Mawatari M, Kukita A. Nupr1 deficiency downregulates HtrA1, enhances SMAD1 signaling, and suppresses age-related bone loss in male mice. J Cell Physiol 2023; 238:566-581. [PMID: 36715607 DOI: 10.1002/jcp.30949] [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: 10/03/2022] [Revised: 12/14/2022] [Accepted: 01/04/2023] [Indexed: 01/31/2023]
Abstract
Nuclear protein 1 (NUPR1) is a stress-induced protein activated by various stresses, such as inflammation and oxidative stress. We previously reported that Nupr1 deficiency increased bone volume by enhancing bone formation in 11-week-old mice. Analysis of differentially expressed genes between wild-type (WT) and Nupr1-knockout (Nupr1-KO) osteocytes revealed that high temperature requirement A 1 (HTRA1), a serine protease implicated in osteogenesis and transforming growth factor-β signaling was markedly downregulated in Nupr1-KO osteocytes. Nupr1 deficiency also markedly reduced HtrA1 expression, but enhanced SMAD1 signaling in in vitro-cultured primary osteoblasts. In contrast, Nupr1 overexpression enhanced HtrA1 expression in osteoblasts, suggesting that Nupr1 regulates HtrA1 expression, thereby suppressing osteoblastogenesis. Since HtrA1 is also involved in cellular senescence and age-related diseases, we analyzed aging-related bone loss in Nupr1-KO mice. Significant spine trabecular bone loss was noted in WT male and female mice during 6-19 months of age, whereas aging-related trabecular bone loss was attenuated, especially in Nupr1-KO male mice. Moreover, cellular senescence-related markers were upregulated in the osteocytes of 6-19-month-old WT male mice but markedly downregulated in the osteocytes of 19-month-old Nupr1-KO male mice. Oxidative stress-induced cellular senescence stimulated Nupr1 and HtrA1 expression in in vitro-cultured primary osteoblasts, and Nupr1 overexpression enhanced p16ink4a expression in osteoblasts. Finally, NUPR1 expression in osteocytes isolated from the bones of patients with osteoarthritis was correlated with age. Collectively, these results indicate that Nupr1 regulates HtrA1-mediated osteoblast differentiation and senescence. Our findings unveil a novel Nupr1/HtrA1 axis, which may play pivotal roles in bone formation and age-related bone loss.
Collapse
Affiliation(s)
- Masatoshi Murayama
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Hirohito Hirata
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Makoto Shiraki
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U 1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology & Oral Anatomy, Kyushu University Graduate School of Dental Science, Fukuoka, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology & Oral Anatomy, Kyushu University Graduate School of Dental Science, Fukuoka, Japan
| | - Toshihisa Komori
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Science, Nagasaki, Japan
| | - Takeshi Moriishi
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Science, Nagasaki, Japan
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Tadatsugu Morimoto
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaaki Mawatari
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Akiko Kukita
- Research Center of Arthroplasty, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
46
|
Targeting NRF2 to promote epithelial repair. Biochem Soc Trans 2023; 51:101-111. [PMID: 36762597 PMCID: PMC9987932 DOI: 10.1042/bst20220228] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/11/2023]
Abstract
The transcription factor NRF2 is well known as a master regulator of the cellular stress response. As such, activation of NRF2 has gained widespread attention for its potential to prevent tissue injury, but also as a possible therapeutic approach to promote repair processes. While NRF2 activation affects most or even all cell types, its effect on epithelial cells during repair processes has been particularly well studied. In response to tissue injury, these cells proliferate, migrate and/or spread to effectively repair the damage. In this review, we discuss how NRF2 governs repair of epithelial tissues, and we highlight the increasing number of NRF2 targets with diverse roles in regulating epithelial repair.
Collapse
|
47
|
Dodson M, Shakya A, Chen J, Chen WT, McKee NW, Zhang DD. The NRF2-anti-ferroptosis Axis in Health and Disease. FERROPTOSIS IN HEALTH AND DISEASE 2023:213-239. [DOI: 10.1007/978-3-031-39171-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|