1
|
Yu H, Ren K, Jin Y, Zhang L, Liu H, Huang Z, Zhang Z, Chen X, Yang Y, Wei Z. Mitochondrial DAMPs: Key mediators in neuroinflammation and neurodegenerative disease pathogenesis. Neuropharmacology 2025; 264:110217. [PMID: 39557152 DOI: 10.1016/j.neuropharm.2024.110217] [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: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are increasingly linked to mitochondrial dysfunction and neuroinflammation. Central to this link are mitochondrial damage-associated molecular patterns (mtDAMPs), including mitochondrial DNA, ATP, and reactive oxygen species, released during mitochondrial stress or damage. These mtDAMPs activate inflammatory pathways, such as the NLRP3 inflammasome and cGAS-STING, contributing to the progression of neurodegenerative diseases. This review delves into the mechanisms by which mtDAMPs drive neuroinflammation and discusses potential therapeutic strategies targeting these pathways to mitigate neurodegeneration. Additionally, it explores the cross-talk between mitochondria and the immune system, highlighting the complex interplay that exacerbates neuronal damage. Understanding the role of mtDAMPs could pave the way for novel treatments aimed at modulating neuroinflammation and slowing disease progression, ultimately improving patient outcome.
Collapse
Affiliation(s)
- Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Zhen Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Ziheng Zhang
- College of Life Sciences, Xinjiang University, Urumqi, Xinjiang, 830046, PR China
| | - Xing Chen
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
2
|
Yu S, Lu X, Li C, Han Z, Li Y, Zhang X, Guo D. TFAM and Mitochondrial Protection in Diabetic Kidney Disease. Diabetes Metab Syndr Obes 2024; 17:4355-4365. [PMID: 39588133 PMCID: PMC11586499 DOI: 10.2147/dmso.s487815] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
Diabetic kidney disease (DKD) is a significant complication of diabetes and a major cause of end-stage renal disease. Affecting around 40% of diabetic patients, DKD poses substantial economic burdens due to its prevalence worldwide. The primary clinical features of DKD include the leakage of proteins into the urine, altered glomerular filtration, and an increased risk of cardiovascular diseases. Current treatments focus on managing hypertension and hyperglycemia to slow the progression of DKD. These include the use of SGLT2 inhibitors to control blood sugar and ACE inhibitors to reduce blood pressure. Despite these measures, current treatments do not cure DKD and fail to address its underlying causes. Emerging research highlights mitochondrial dysfunction as a pivotal factor in DKD progression. The kidneys' high energy requirements make them particularly susceptible to disturbances in mitochondrial function. In DKD, mitochondrial damage leads to reduced energy production and increased oxidative stress, exacerbating tissue damage. Mitochondrial DNA (mtDNA) damage is a key aspect of this dysfunction, with studies suggesting that changes in mtDNA copy number can serve as biomarkers for the progression of the disease. Efforts to target mitochondrial dysfunction are gaining traction as a potential therapeutic strategy. This includes promoting mitochondrial health through pharmacological and lifestyle interventions aimed at enhancing mitochondrial function and reducing oxidative stress. Such approaches could lead to more effective treatments that directly address the DKD.
Collapse
Affiliation(s)
- Siming Yu
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, People’s Republic of China
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xinxin Lu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Chunsheng Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zehui Han
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yue Li
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xianlong Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Dandan Guo
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, People’s Republic of China
| |
Collapse
|
3
|
Cao H, Xiao J, Baylink DJ, Nguyen V, Shim N, Lee J, Mallari DJR, Wasnik S, Mirshahidi S, Chen CS, Abdel-Azim H, Reeves ME, Xu Y. Development of a Competitive Nutrient-Based T-Cell Immunotherapy Designed to Block the Adaptive Warburg Effect in Acute Myeloid Leukemia. Biomedicines 2024; 12:2250. [PMID: 39457563 PMCID: PMC11504511 DOI: 10.3390/biomedicines12102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: T-cell-based adoptive cell therapies have emerged at the forefront of cancer immunotherapies; however, failed long-term survival and inevitable exhaustion of transplanted T lymphocytes in vivo limits clinical efficacy. Leukemia blasts possess enhanced glycolysis (Warburg effect), exploiting their microenvironment to deprive nutrients (e.g., glucose) from T cells, leading to T-cell dysfunction and leukemia progression. Methods: Thus, we explored whether genetic reprogramming of T-cell metabolism could improve their survival and empower T cells with a competitive glucose-uptake advantage against blasts and inhibit their uncontrolled proliferation. Results: Here, we discovered that high-glucose concentration reduced the T-cell expression of glucose transporter GLUT1 (SLC2A1) and TFAM (mitochondrion transcription factor A), an essential transcriptional regulator of mitochondrial biogenesis, leading to their impaired expansion ex vivo. To overcome the glucose-induced genetic deficiency in metabolism, we engineered T cells with lentiviral overexpression of SLC2A1 and/or TFAM transgene. Multi-omics analyses revealed that metabolic reprogramming promoted T-cell proliferation by increasing IL-2 release and reducing exhaustion. Moreover, the engineered T cells competitively deprived glucose from allogenic blasts and lessened leukemia burden in vitro. Conclusions: Our findings propose a novel T-cell immunotherapy that utilizes a dual strategy of starving blasts and cytotoxicity for preventing uncontrolled leukemia proliferation.
Collapse
Affiliation(s)
- Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Vinh Nguyen
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Nathan Shim
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jae Lee
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Dave J. R. Mallari
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Saied Mirshahidi
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Biospecimen Laboratory, Department of Medicine and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hisham Abdel-Azim
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Transplant and Cell Therapy, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
- Division of Hematology and Oncology, Department of Pediatrics, Loma Linda University, Loma Linda, CA 92354, USA
| | - Mark E. Reeves
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yi Xu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
4
|
Li Y, Tian L, Li S, Chen X, Lei F, Bao J, Wu Q, Wen Y, Jie Y. Disrupted mitochondrial transcription factor A expression promotes mitochondrial dysfunction and enhances ocular surface inflammation by activating the absent in melanoma 2 inflammasome. Free Radic Biol Med 2024; 222:106-121. [PMID: 38797339 DOI: 10.1016/j.freeradbiomed.2024.05.032] [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: 03/28/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE Severe dry eye disease causes ocular surface damage, which is highly associated with mitochondrial dysfunction. Mitochondrial transcription factor A (TFAM) is essential for packaging mitochondrial DNA (mtDNA) and is crucial for maintaining mitochondrial function. Herein, we aimed to explore the effect of a decreased TFAM expression on ocular surface damage. METHODS Female C57BL/6 mice were induced ocular surface injury by topical administrating benzalkonium chloride (BAC). Immortalized human corneal epithelial cells (HCECs) were stimulated by tert-butyl hydroperoxide (t-BHP) to create oxidative stress damage. HCECs with TFAM knockdown were established. RNA sequencing was employed to analyze the whole-genome expression. Mitochondrial changes were measured by transmission electron microscopy, Seahorse metabolic flux analysis, mitochondrial membrane potential, and mtDNA copy number. TFAM expression and inflammatory cytokines were determined using RT-qPCR, immunohistochemistry, immunofluorescence, and immunoblotting. RESULTS In both the corneas of BAC-treated mice and t-BHP-induced HCECs, we observed impaired TFAM expression, accompanied by mitochondrial structure and function defects. TFAM downregulation in HCECs suppressed mitochondrial respiratory capacity, reduced mtDNA content, induced mtDNA leakage into the cytoplasm, and led to inflammation. RNA sequencing revealed the absent in melanoma 2 (AIM2) inflammasome was activated in the corneas of BAC-treated mice. The AIM2 inflammasome activation was confirmed in TFAM knockdown HCECs. TFAM knockdown in t-BHP-stimulated HCECs aggravated mitochondrial dysfunction and the AIM2 inflammasome activation, thereby further triggering the secretion of inflammatory factors such as interleukin (IL) -1β and IL-18. CONCLUSIONS TFAM reduction impaired mitochondrial function, activated AIM2 inflammasome and promoted ocular surface inflammation, revealing an underlying molecular mechanism for ocular surface disorders.
Collapse
Affiliation(s)
- Yaqiong Li
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Lei Tian
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Siyuan Li
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Xiaoniao Chen
- Department of Ophthalmology, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China.
| | - Fengyang Lei
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Jiayu Bao
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Qianru Wu
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Ya Wen
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| | - Ying Jie
- Beijing Institute of Ophthalmology, Beijing TongRen Eye Center, Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| |
Collapse
|
5
|
Song Y, Wang W, Wang B, Shi Q. The Protective Mechanism of TFAM on Mitochondrial DNA and its Role in Neurodegenerative Diseases. Mol Neurobiol 2024; 61:4381-4390. [PMID: 38087167 DOI: 10.1007/s12035-023-03841-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/28/2023] [Indexed: 07/11/2024]
Abstract
Mitochondrial transcription factor A (TFAM) is a mitochondrial protein encoded by nuclear genes and transported from the cytoplasm to the mitochondria. TFAM is essential for the maintenance, expression, and delivery of mitochondrial DNA (mtDNA) and can regulate the replication and transcription of mtDNA. TFAM is associated with the formation of mtDNA nucleomimetic structures, mtDNA repair, and mtDNA stability. However, the mechanism by which TFAM protects mtDNA is still being studied. This review provides a summary of the protective mechanism of TFAM on mtDNA including the discrete regulatory effects of TFAM acetylation and phosphorylation on mtDNA, the regulation of Ca2+ levels by TFAM to activate transcription in mitochondria, and the increased binding of TFAM to mtDNA damage hot spots. This review also discusses the association between TFAM and some neurodegenerative diseases.
Collapse
Affiliation(s)
- Ying Song
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People's Republic of China.
- Hangzhou King's Bio-Pharmaceutical Technology Co., Ltd., Hangzhou, 310007, Zhejiang, China.
| | - Wenjun Wang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People's Republic of China
| | - Beibei Wang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People's Republic of China
| | - Qiwen Shi
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People's Republic of China
| |
Collapse
|
6
|
Zhang Y, Wei S, Jin EJ, Jo Y, Oh CM, Bae GU, Kang JS, Ryu D. Protein Arginine Methyltransferases: Emerging Targets in Cardiovascular and Metabolic Disease. Diabetes Metab J 2024; 48:487-502. [PMID: 39043443 PMCID: PMC11307121 DOI: 10.4093/dmj.2023.0362] [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: 10/11/2023] [Accepted: 06/17/2024] [Indexed: 07/25/2024] Open
Abstract
Cardiovascular diseases (CVDs) and metabolic disorders stand as formidable challenges that significantly impact the clinical outcomes and living quality for afflicted individuals. An intricate comprehension of the underlying mechanisms is paramount for the development of efficacious therapeutic strategies. Protein arginine methyltransferases (PRMTs), a class of enzymes responsible for the precise regulation of protein methylation, have ascended to pivotal roles and emerged as crucial regulators within the intrinsic pathophysiology of these diseases. Herein, we review recent advancements in research elucidating on the multifaceted involvements of PRMTs in cardiovascular system and metabolic diseases, contributing significantly to deepen our understanding of the pathogenesis and progression of these maladies. In addition, this review provides a comprehensive analysis to unveil the distinctive roles of PRMTs across diverse cell types implicated in cardiovascular and metabolic disorders, which holds great potential to reveal novel therapeutic interventions targeting PRMTs, thus presenting promising perspectives to effectively address the substantial global burden imposed by CVDs and metabolic disorders.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, Korea
| | - Shibo Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Eun-Ju Jin
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Gyu-Un Bae
- Muscle Physiome Institute, College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
- Research Institute of Aging-Related Diseases, AniMusCure Inc., Suwon, Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, Korea
- Research Institute of Aging-Related Diseases, AniMusCure Inc., Suwon, Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
7
|
Zhou Y, Huang X, Jin Y, Qiu M, Ambe PC, Basharat Z, Hong W. The role of mitochondrial damage-associated molecular patterns in acute pancreatitis. Biomed Pharmacother 2024; 175:116690. [PMID: 38718519 DOI: 10.1016/j.biopha.2024.116690] [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: 02/08/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 06/03/2024] Open
Abstract
Acute pancreatitis (AP) is one of the most common gastrointestinal tract diseases with significant morbidity and mortality. Current treatments remain unspecific and supportive due to the severity and clinical course of AP, which can fluctuate rapidly and unpredictably. Mitochondria, cellular power plant to produce energy, are involved in a variety of physiological or pathological activities in human body. There is a growing evidence indicating that mitochondria damage-associated molecular patterns (mtDAMPs) play an important role in pathogenesis and progression of AP. With the pro-inflammatory properties, released mtDAMPs may damage pancreatic cells by binding with receptors, activating downstream molecules and releasing inflammatory factors. This review focuses on the possible interaction between AP and mtDAMPs, which include cytochrome c (Cyt c), mitochondrial transcription factor A (TFAM), mitochondrial DNA (mtDNA), cardiolipin (CL), adenosine triphosphate (ATP) and succinate, with focus on experimental research and potential therapeutic targets in clinical practice. Preventing or diminishing the release of mtDAMPs or targeting the mtDAMPs receptors might have a role in AP progression.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Xiaoyi Huang
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yinglu Jin
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China; School of the First Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Minhao Qiu
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Peter C Ambe
- Department of General Surgery, Visceral Surgery and Coloproctology, Vinzenz-Pallotti-Hospital Bensberg, Vinzenz-Pallotti-Str. 20-24, Bensberg 51429, Germany
| | | | - Wandong Hong
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
8
|
Liu H, Zhen C, Xie J, Luo Z, Zeng L, Zhao G, Lu S, Zhuang H, Fan H, Li X, Liu Z, Lin S, Jiang H, Chen Y, Cheng J, Cao Z, Dai K, Shi J, Wang Z, Hu Y, Meng T, Zhou C, Han Z, Huang H, Zhou Q, He P, Feng D. TFAM is an autophagy receptor that limits inflammation by binding to cytoplasmic mitochondrial DNA. Nat Cell Biol 2024; 26:878-891. [PMID: 38783142 DOI: 10.1038/s41556-024-01419-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/08/2024] [Indexed: 05/25/2024]
Abstract
When cells are stressed, DNA from energy-producing mitochondria can leak out and drive inflammatory immune responses if not cleared. Cells employ a quality control system called autophagy to specifically degrade damaged components. We discovered that mitochondrial transcription factor A (TFAM)-a protein that binds mitochondrial DNA (mtDNA)-helps to eliminate leaked mtDNA by interacting with the autophagy protein LC3 through an autolysosomal pathway (we term this nucleoid-phagy). TFAM contains a molecular zip code called the LC3 interacting region (LIR) motif that enables this binding. Although mutating TFAM's LIR motif did not affect its normal mitochondrial functions, more mtDNA accumulated in the cell cytoplasm, activating inflammatory signalling pathways. Thus, TFAM mediates autophagic removal of leaked mtDNA to restrict inflammation. Identifying this mechanism advances understanding of how cells exploit autophagy machinery to selectively target and degrade inflammatory mtDNA. These findings could inform research on diseases involving mitochondrial damage and inflammation.
Collapse
Affiliation(s)
- Hao Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Huaihe Hospital of Henan University, Kaifeng City, China
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Cien Zhen
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Biology, University of Padova, Padova, Italy
| | - Jianming Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhenhuan Luo
- Department of Cardiology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Lin Zeng
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guojun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Shaohua Lu
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Haixia Zhuang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hualin Fan
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Biology, University of Padova, Padova, Italy
| | - Xia Li
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhaojie Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shiyin Lin
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huilin Jiang
- Emergency Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqian Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jiahao Cheng
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Zhiyu Cao
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The First Clinical Medical School, Guangzhou Medical University, Guangzhou, China
| | - Keyu Dai
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jinhua Shi
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhaohua Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yongquan Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tian Meng
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Chuchu Zhou
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhiyuan Han
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huansen Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qinghua Zhou
- Department of Cardiology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Pengcheng He
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Cardiology, Heyuan People's Hospital, Heyuan, China
| | - Du Feng
- State Key Laboratory of Respiratory Disease, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China.
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Wang V, Tseng KY, Kuo TT, Huang EYK, Lan KL, Chen ZR, Ma KH, Greig NH, Jung J, Choi HI, Olson L, Hoffer BJ, Chen YH. Attenuating mitochondrial dysfunction and morphological disruption with PT320 delays dopamine degeneration in MitoPark mice. J Biomed Sci 2024; 31:38. [PMID: 38627765 PMCID: PMC11022395 DOI: 10.1186/s12929-024-01025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Mitochondria are essential organelles involved in cellular energy production. Changes in mitochondrial function can lead to dysfunction and cell death in aging and age-related disorders. Recent research suggests that mitochondrial dysfunction is closely linked to neurodegenerative diseases. Glucagon-like peptide-1 receptor (GLP-1R) agonist has gained interest as a potential treatment for Parkinson's disease (PD). However, the exact mechanisms responsible for the therapeutic effects of GLP-1R-related agonists are not yet fully understood. METHODS In this study, we explores the effects of early treatment with PT320, a sustained release formulation of the GLP-1R agonist Exenatide, on mitochondrial functions and morphology in a progressive PD mouse model, the MitoPark (MP) mouse. RESULTS Our findings demonstrate that administration of a clinically translatable dose of PT320 ameliorates the reduction in tyrosine hydroxylase expression, lowers reactive oxygen species (ROS) levels, and inhibits mitochondrial cytochrome c release during nigrostriatal dopaminergic denervation in MP mice. PT320 treatment significantly preserved mitochondrial function and morphology but did not influence the reduction in mitochondria numbers during PD progression in MP mice. Genetic analysis indicated that the cytoprotective effect of PT320 is attributed to a reduction in the expression of mitochondrial fission protein 1 (Fis1) and an increase in the expression of optic atrophy type 1 (Opa1), which is known to play a role in maintaining mitochondrial homeostasis and decreasing cytochrome c release through remodeling of the cristae. CONCLUSION Our findings suggest that the early administration of PT320 shows potential as a neuroprotective treatment for PD, as it can preserve mitochondrial function. Through enhancing mitochondrial health by regulating Opa1 and Fis1, PT320 presents a new neuroprotective therapy in PD.
Collapse
Affiliation(s)
- Vicki Wang
- Doctoral Degree Program in Translational Medicine, National Defense Medical Center and Academia Sinica, Taipei, 11490, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Kuan-Yin Tseng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, 11490, Taiwan
- National Defense Medical Center, Taipei, 11490, Taiwan
| | - Tung-Tai Kuo
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, 11490, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Kuo-Lun Lan
- Department of Pathology, Tri-Service General Hospital, Taipei, 11490, Taiwan
| | - Zi-Rong Chen
- Department of Pathology, Tri-Service General Hospital, Taipei, 11490, Taiwan
| | - Kuo-Hsing Ma
- Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD, 21224, USA
| | - Jin Jung
- Peptron, Inc., Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Ho-Ii Choi
- Peptron, Inc., Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, 171 77, Stockholm, Sweden
| | - Barry J Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, 11490, Taiwan.
- National Defense Medical Center, Taipei, 11490, Taiwan.
| |
Collapse
|
10
|
Kobayashi H, Yoshimoto C, Matsubara S, Shigetomi H, Imanaka S. Altered Energy Metabolism, Mitochondrial Dysfunction, and Redox Imbalance Influencing Reproductive Performance in Granulosa Cells and Oocyte During Aging. Reprod Sci 2024; 31:906-916. [PMID: 37917297 DOI: 10.1007/s43032-023-01394-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Female fertility decreases during aging. The development of effective therapeutic strategies to address the age-related decline in oocyte quality and quantity and its accurate diagnosis remain major challenges. In this review, we summarize our current understanding of the study of aging and infertility, focusing primarily on the molecular basis of energy metabolism, mitochondrial function, and redox homeostasis in granulosa cells and oocytes, and discuss perspectives on future research directions. Mitochondria serve as a central hub sensing a multitude of physiological processes, including energy production, cellular redox homeostasis, aging, and senescence. Young granulosa cells favor glycolysis and actively produce pyruvate, NADPH, and other metabolites. Oocytes rely on oxidative phosphorylation fueled by nutrients, metabolites, and antioxidants provided by the adjacent granulosa cells. A reduced cellular energy metabolism phenotype, including both aerobic glycolysis and mitochondrial respiration, is characteristic of older female granulosa cells compared with younger female granulosa cells. Aged oocytes become more susceptible to oxidative damage to cells and mitochondria because of further depletion of antioxidant-dependent ROS scavenging systems. Molecular perturbations of gene expression caused by a subtle change in the follicular fluid microenvironment adversely affect energy metabolism and mitochondrial dynamics in granulosa cells and oocytes, further causing redox imbalance and accelerating aging and senescence. Furthermore, recent advances in technology are beginning to identify biofluid molecular markers that may influence follicular development and oocyte quality. Accumulating evidence suggests that redox imbalance caused by abnormal energy metabolism and/or mitochondrial dysfunction is closely linked to the pathophysiology of age-related subfertility.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-Cho, Kashihara, 634-0813, Japan.
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan.
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, 2-897-5 Shichi-Jyonishi-Machi, Nara, 630-8581, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan
- Department of Medicine, Kei Oushin Clinic, 5-2-6, Naruo-Cho, Nishinomiya, 663-8184, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, 3-3-17 Kitatomigaoka-Cho, Nara, 634-0001, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-Cho, Kashihara, 634-0813, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, 634-8522, Japan
| |
Collapse
|
11
|
Torp MK, Stensløkken KO, Vaage J. When Our Best Friend Becomes Our Worst Enemy: The Mitochondrion in Trauma, Surgery, and Critical Illness. J Intensive Care Med 2024:8850666241237715. [PMID: 38505947 DOI: 10.1177/08850666241237715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Common for major surgery, multitrauma, sepsis, and critical illness, is a whole-body inflammation. Tissue injury is able to trigger a generalized inflammatory reaction. Cell death causes release of endogenous structures termed damage associated molecular patterns (DAMPs) that initiate a sterile inflammation. Mitochondria are evolutionary endosymbionts originating from bacteria, containing molecular patterns similar to bacteria. These molecular patterns are termed mitochondrial DAMPs (mDAMPs). Mitochondrial debris released into the extracellular space or into the circulation is immunogenic and damaging secondary to activation of the innate immune system. In the circulation, released mDAMPS are either free or exist in extracellular vesicles, being able to act on every organ and cell in the body. However, the role of mDAMPs in trauma and critical care is not fully clarified. There is a complete lack of knowledge how they may be counteracted in patients. Among mDAMPs are mitochondrial DNA, cardiolipin, N-formyl peptides, cytochrome C, adenosine triphosphate, reactive oxygen species, succinate, and mitochondrial transcription factor A. In this overview, we present the different mDAMPs, their function, release, targets, and inflammatory potential. In light of present knowledge, the role of mDAMPs in the pathophysiology of major surgery and trauma as well as sepsis, and critical care is discussed.
Collapse
Affiliation(s)
- May-Kristin Torp
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
- Department of Research, Østfold Hospital Trust, Grålum, Norway
| | - Kåre-Olav Stensløkken
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
| | - Jarle Vaage
- Section of Physiology, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, Oslo, Norway
- Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Zhao B, Zhang Q, He Y, Cao W, Song W, Liang X. Targeted metabolomics reveals the aberrant energy status in diabetic peripheral neuropathy and the neuroprotective mechanism of traditional Chinese medicine JinMaiTong. J Pharm Anal 2024; 14:225-243. [PMID: 38464790 PMCID: PMC10921333 DOI: 10.1016/j.jpha.2023.09.007] [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: 06/15/2023] [Revised: 08/15/2023] [Accepted: 09/18/2023] [Indexed: 03/12/2024] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a common and devastating complication of diabetes, for which effective therapies are currently lacking. Disturbed energy status plays a crucial role in DPN pathogenesis. However, the integrated profile of energy metabolism, especially the central carbohydrate metabolism, remains unclear in DPN. Here, we developed a metabolomics approach by targeting 56 metabolites using high-performance ion chromatography-tandem mass spectrometry (HPIC-MS/MS) to illustrate the integrative characteristics of central carbohydrate metabolism in patients with DPN and streptozotocin-induced DPN rats. Furthermore, JinMaiTong (JMT), a traditional Chinese medicine (TCM) formula, was found to be effective for DPN, improving the peripheral neurological function and alleviating the neuropathology of DPN rats even after demyelination and axonal degeneration. JMT ameliorated DPN by regulating the aberrant energy balance and mitochondrial functions, including excessive glycolysis restoration, tricarboxylic acid cycle improvement, and increased adenosine triphosphate (ATP) generation. Bioenergetic profile was aberrant in cultured rat Schwann cells under high-glucose conditions, which was remarkably corrected by JMT treatment. In-vivo and in-vitro studies revealed that these effects of JMT were mainly attributed to the activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) and downstream peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Our results expand the therapeutic framework for DPN and suggest the integrative modulation of energy metabolism using TCMs, such as JMT, as an effective strategy for its treatment.
Collapse
Affiliation(s)
- Bingjia Zhao
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qian Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yiqian He
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weifang Cao
- Institute of Basic Medicine Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Wei Song
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaochun Liang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
13
|
Wang Y, Wang J, Tao SY, Liang Z, Xie R, Liu NN, Deng R, Zhang Y, Deng D, Jiang G. Mitochondrial damage-associated molecular patterns: A new insight into metabolic inflammation in type 2 diabetes mellitus. Diabetes Metab Res Rev 2024; 40:e3733. [PMID: 37823338 DOI: 10.1002/dmrr.3733] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/18/2023] [Accepted: 09/08/2023] [Indexed: 10/13/2023]
Abstract
The pathogenesis of diabetes is accompanied by increased levels of inflammatory factors, also known as "metabolic inflammation", which runs through the whole process of the occurrence and development of the disease. Mitochondria, as the key site of glucose and lipid metabolism, is often accompanied by mitochondrial function damage in type 2 diabetes mellitus (T2DM). Damaged mitochondria release pro-inflammatory factors through damage-related molecular patterns that activate inflammation pathways and reactions to oxidative stress, further aggravate metabolic disorders, and form a vicious circle. Currently, the pathogenesis of diabetes is still unclear, and clinical treatment focuses primarily on symptomatic intervention of the internal environment of disorders of glucose and lipid metabolism with limited clinical efficacy. The proinflammatory effect of mitochondrial damage-associated molecular pattern (mtDAMP) in T2DM provides a new research direction for exploring the pathogenesis and intervention targets of T2DM. Therefore, this review covers the most recent findings on the molecular mechanism and related signalling cascades of inflammation caused by mtDAMP in T2DM and discusses its pathogenic role of it in the pathological process of T2DM to search potential intervention targets.
Collapse
Affiliation(s)
- Yan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jingwu Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Si-Yu Tao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | | | - Rong Xie
- Xinjiang Medical University, Urumqi, China
| | - Nan-Nan Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ruxue Deng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuelin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Deqiang Deng
- Department of Endocrinology, Urumqi Hospital of Traditional Chinese Medicine, Urumqi, China
| | - Guangjian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Han D, Yang N, Liu H, Yao Y, Xu S. TBBPA causes apoptosis in grass carp hepatocytes involving destroyed ER-mitochondrial function. CHEMOSPHERE 2023; 341:139974. [PMID: 37648165 DOI: 10.1016/j.chemosphere.2023.139974] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
Tetrabromobisphenol A (TBBPA) is the most-produced brominated flame retardant, which can be found in various industrial and household products. Studies have shown that TBBPA has hepatotoxicity, and could pose a risk to aquatic animals. The endoplasmic reticulum (ER) and mitochondria are two important organelles that are highly dynamic in cells, the homeostasis and orchestrated interactions of which are crucial to maintaining cellular function. The aim of this study was to explore the involvement of ER-mitochondria crosstalk in TBBPA-induced toxicity in aquatic animals' hepatocytes. Herein, we exposed grass carp hepatocytes (L8824 cells) to different concentrations of TBBPA. Our experimental results suggested that TBBPA exposure suppressed cell viability and caused apoptosis of L8824 cells. TBBPA treatment upregulated expressions of ER stress markers, increased reactive oxygen species (ROS) and mitochondrial Ca2+ levels, and reduced mitochondrial membrane potential (MMP) in L8824 cells. However, the pretreatment of 2-aminoethoxydiphenyl borate (2-APB) could alleviate TBBPA-induced cell apoptosis, ER stress, and mitochondrial dysfunction. Additionally, 2-APB pretreat relieved ER-mitochondrial contact and the expression of ER-mitochondrial function-related genes induced by high-dose TBBPA. Taken together, these results indicated that TBBPA caused grass carp hepatocyte apoptosis by destroying ER-mitochondrial crosstalk.
Collapse
Affiliation(s)
- Dongxu Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Naixi Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Huanyi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yujie Yao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
15
|
Hsu YL, Chen HJ, Gao JX, Yang MY, Fu RH. Chiisanoside Mediates the Parkin/ZNF746/PGC-1α Axis by Downregulating MiR-181a to Improve Mitochondrial Biogenesis in 6-OHDA-Caused Neurotoxicity Models In Vitro and In Vivo: Suggestions for Prevention of Parkinson's Disease. Antioxidants (Basel) 2023; 12:1782. [PMID: 37760085 PMCID: PMC10525196 DOI: 10.3390/antiox12091782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The degeneration of dopamine (DA) neurons is known to be associated with defects in mitochondrial biogenesis caused by aging, environmental factors, or mutations in genes, leading to Parkinson's disease (PD). As PD has not yet been successfully cured, the strategy of using small molecule drugs to protect and restore mitochondrial biogenesis is a promising direction. This study evaluated the efficacy of synthetic chiisanoside (CSS) identified in the leaves of Acanthopanax sessiliflorus to prevent PD symptoms. The results show that in the 6-hydroxydopamine (6-OHDA) model, CSS pretreatment can effectively alleviate the reactive oxygen species generation and apoptosis of SH-SY5Y cells, thereby lessening the defects in the C. elegans model including DA neuron degeneration, dopamine-mediated food sensitivity behavioral disorders, and shortened lifespan. Mechanistically, we found that CSS could restore the expression of proliferator-activated receptor gamma coactivator-1-alpha (PGC-1α), a key molecule in mitochondrial biogenesis, and its downstream related genes inhibited by 6-OHDA. We further confirmed that this is due to the enhanced activity of parkin leading to the ubiquitination and degradation of PGC-1α inhibitor protein Zinc finger protein 746 (ZNF746). Parkin siRNA treatment abolished this effect of CSS. Furthermore, we found that CSS inhibited 6-OHDA-induced expression of miR-181a, which targets parkin. The CSS's ability to reverse the 6-OHDA-induced reduction in mitochondrial biogenesis and activation of apoptosis was abolished after the transfection of anti-miR-181a and miR-181a mimics. Therefore, the neuroprotective effect of CSS mainly promotes mitochondrial biogenesis by regulating the miR-181a/Parkin/ZNF746/PGC-1α axis. CSS potentially has the opportunity to be developed into PD prevention agents.
Collapse
Affiliation(s)
- Yu-Ling Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-L.H.); (H.-J.C.); (J.-X.G.)
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Hui-Jye Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-L.H.); (H.-J.C.); (J.-X.G.)
| | - Jia-Xin Gao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-L.H.); (H.-J.C.); (J.-X.G.)
| | - Ming-Yang Yang
- Ph.D. Program for Aging, China Medical University, Taichung 40402, Taiwan;
| | - Ru-Huei Fu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-L.H.); (H.-J.C.); (J.-X.G.)
- Ph.D. Program for Aging, China Medical University, Taichung 40402, Taiwan;
- Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| |
Collapse
|
16
|
Wang J, Chen S, Zhao X, Guo Q, Yang R, Zhang C, Huang Y, Ma L, Zhao S. Effect of PPARγ on oxidative stress in diabetes-related dry eye. Exp Eye Res 2023; 231:109498. [PMID: 37169280 DOI: 10.1016/j.exer.2023.109498] [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: 01/13/2023] [Revised: 04/08/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023]
Abstract
Oxidative stress is closely associated with diabetes and can cause free radical accumulation and eventually lead to ocular surface tissue damage. The purpose of this study was to investigate peroxisome proliferator-activated receptor-γ (PPARγ) expression in the lacrimal gland (LG), meibomian gland, and cornea of diabetes-related dry eye mice and whether the PPARγ agonist rosiglitazone can alleviate the oxidative stress of the ocular surface, thereby improving the condition of diabetes-related dry eye. Quantitative RT-PCR (Q-PCR) showed that the PPARγ, catalase, glutathione peroxidase 3, and heme oxygenase-1 (HO-1) mRNA expression levels in the LG of diabetes-related dry eye mice decreased at 8 and 12 weeks. In addition, the increased levels of oxidative stress were confirmed by western blot. Although the mRNA expression levels of antioxidant enzymes in the cornea and meibomian gland decreased at 8 weeks, some of them recovered by 12 weeks. Rosiglitazone alleviated ocular surface damage and increased corneal sensitivity and tear production in diabetes-related dry eye mice. Moreover, the reactive oxygen species accumulation was reduced and the PPARγ, HO-1, and glutathione peroxidase 3 mRNA expression levels were increased in the LG. The PPARγ, HO-1, translocase of the outer membrane 20, and mitochondrial transcription factor A protein levels were also significantly increased. These results demonstrated that rosiglitazone reduced oxidative stress in the LG of diabetes-related dry eye mice, at least in part, by activating PPARγ to up-regulate antioxidant enzyme expression.
Collapse
Affiliation(s)
- Jing Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Shuangping Chen
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xiuxiu Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China; Department of Ophthalmology, Tianjin Prevention and Treatment Center for Occupational Diseases, Tianjin, 300011, China
| | - Qian Guo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Ruibo Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Chen Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Yue Huang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Lechong Ma
- Department of Molecular Cell and Department Biology, California State University Los Angeles, Los Angeles, CA, USA
| | - Shaozhen Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China.
| |
Collapse
|
17
|
Protective Effect of Ergothioneine against 7-Ketocholesterol-Induced Mitochondrial Damage in hCMEC/D3 Human Brain Endothelial Cells. Int J Mol Sci 2023; 24:ijms24065498. [PMID: 36982572 PMCID: PMC10056831 DOI: 10.3390/ijms24065498] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Recent findings have suggested that the natural compound ergothioneine (ET), which is synthesised by certain fungi and bacteria, has considerable cytoprotective potential. We previously demonstrated the anti-inflammatory effects of ET on 7-ketocholesterol (7KC)-induced endothelial injury in human blood-brain barrier endothelial cells (hCMEC/D3). 7KC is an oxidised form of cholesterol present in atheromatous plaques and the sera of patients with hypercholesterolaemia and diabetes mellitus. The aim of this study was to elucidate the protective effect of ET on 7KC-induced mitochondrial damage. Exposure of human brain endothelial cells to 7KC led to a loss of cell viability, together with an increase in intracellular free calcium levels, increased cellular and mitochondrial reactive oxygen species, a decrease in mitochondrial membrane potential, reductions in ATP levels, and increases in mRNA expression of TFAM, Nrf2, IL-1β, IL-6 and IL-8. These effects were significantly decreased by ET. Protective effects of ET were diminished when endothelial cells were coincubated with verapamil hydrochloride (VHCL), a nonspecific inhibitor of the ET transporter OCTN1 (SLC22A4). This outcome demonstrates that ET-mediated protection against 7KC-induced mitochondrial damage occurred intracellularly and not through direct interaction with 7KC. OCTN1 mRNA expression itself was significantly increased in endothelial cells after 7KC treatment, consistent with the notion that stress and injury may increase ET uptake. Our results indicate that ET can protect against 7KC-induced mitochondrial injury in brain endothelial cells.
Collapse
|
18
|
gp130 Activates Mitochondrial Dynamics for Hepatocyte Survival in a Model of Steatohepatitis. Biomedicines 2023; 11:biomedicines11020396. [PMID: 36830933 PMCID: PMC9953457 DOI: 10.3390/biomedicines11020396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Obesity is the main cause of metabolic complications. Fatty liver infiltration is a companion of obesity. NAFLD is associated with impaired energy metabolism with an excess of nutrients. Mitochondrial dynamics are important for the regulation of energy balance, which regulates mitochondrial function, apoptosis, and mitophagy. The aim of this study was to investigate the effect of gp130 on the components of mitochondrial dynamics in a cellular model of steatohepatitis. Addition of IL-6/gp130 contributed to an increase in the percentage of live cells and a decrease in the percentage of dead and apoptotic cells. Addition of IL-6/gp130 increased the expression of NF-kB1 gene and mitochondrial dynamics markers (MFN2 and TFAM) in HepG2 with tBHP/Oleic. Addition of IL-6 or gp130 reduced the expression of cytoprotector genes (HSF1 and HSP70) in HepG2 cell cultures with tBHP/Oleic. Increased mitochondrial dynamics gene activity protected against HepG2 cell death in the steatohepatitis model. Trans-signaling resulted in increased TFAM and MAPLC3B, and decreased DNM1L gene expression in HepG2 with tBHP/Oleic.
Collapse
|
19
|
Effects of the anti-inflammatory drug celecoxib on cell death signaling in human colon cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1171-1185. [PMID: 36692829 DOI: 10.1007/s00210-023-02399-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023]
Abstract
The anti-inflammatory drug celecoxib, the only inhibitor of cyclooxygenase-2 (COX-2) with anticancer activity, is used to treat rheumatoid arthritis and can cause endoplasmic reticulum (ER) stress by inhibiting sarco/ER Ca2 +-ATPase activity in cancer cells. This study aimed to investigate the correlation between celecoxib-induced ER stress and the effects of celecoxib against cell death signaling. Treatment of human colon cancer HCT116 cells with celecoxib reduced their viability and resulted in a loss of mitochondrial membrane potential ([Formula: see text]). Additionally, celecoxib treatment reduced the expression of genes involved in mitochondrial biogenesis and metabolism such as mitochondrial transcription factor A (TFAM) and uncoupling protein 2 (UCP2). Furthermore, celecoxib reduced transmembrane protein 117 (TMEM117), and RNAi-mediated knockdown of TMEM117 reduced TFAM and UCP2 expressions. These results suggest that celecoxib treatment results in the loss of [Formula: see text] by reducing TMEM117 expression and provide insights for the development of novel drugs through TMEM117 expression.
Collapse
|
20
|
Pharmacological Activation of Rev-erb α Attenuates Doxorubicin-Induced Cardiotoxicity by PGC-1 α Signaling Pathway. Cardiovasc Ther 2023; 2023:2108584. [PMID: 36874248 PMCID: PMC9977526 DOI: 10.1155/2023/2108584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 02/24/2023] Open
Abstract
Background Doxorubicin-induced cardiotoxicity has been closely concerned in clinical practice. Rev-erbα is a transcriptional repressor that emerges as a drug target for heart diseases recently. This study is aimed at investigating the role and mechanism of Rev-erbα in doxorubicin-induced cardiotoxicity. Methods H9c2 cells were treated with 1.5 μM doxorubicin, and C57BL/6 mice were treated with a 20 mg/kg cumulative dose of doxorubicin to construct doxorubicin-induced cardiotoxicity models in vitro and in vivo. Agonist SR9009 was used to activate Rev-erbα. PGC-1α expression level was downregulated by specific siRNA in H9c2 cells. Cell apoptosis, cardiomyocyte morphology, mitochondrial function, oxidative stress, and signaling pathways were measured. Results SR9009 alleviated doxorubicin-induced cell apoptosis, morphological disorder, mitochondrial dysfunction, and oxidative stress in H9c2 cells and C57BL/6 mice. Meanwhile, PGC-1α and downstream signaling NRF1, TAFM, and UCP2 expression levels were preserved by SR9009 in doxorubicin-treated cardiomyocytes in vitro and in vivo. When downregulating PGC-1α expression level by specific siRNA, the protective role of SR9009 in doxorubicin-treated cardiomyocytes was attenuated with increased cell apoptosis, mitochondrial dysfunction, and oxidative stress. Conclusion Pharmacological activation of Rev-erbα by SR9009 could attenuate doxorubicin-induced cardiotoxicity through preservation of mitochondrial function and alleviation of apoptosis and oxidative stress. The mechanism is associated with the activation of PGC-1α signaling pathways, suggesting that PGC-1α signaling is a mechanism for the protective effect of Rev-erbα against doxorubicin-induced cardiotoxicity.
Collapse
|
21
|
Abad-Jiménez Z, Vezza T, López-Domènech S, Fernández-Reyes M, Canet F, Morillas C, Gómez-Abril SÁ, Bañuls C, Víctor VM, Rocha M. Impact of Roux-en-Y Gastric Bypass on Mitochondrial Biogenesis and Dynamics in Leukocytes of Obese Women. Antioxidants (Basel) 2022; 11:antiox11071302. [PMID: 35883794 PMCID: PMC9312345 DOI: 10.3390/antiox11071302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
The chronic low-grade inflammation widely associated with obesity can lead to a prooxidant status that triggers mitochondrial dysfunction. To date, Roux-en-Y gastric bypass (RYGB) is considered the most effective strategy for obese patients. However, little is known about its molecular mechanisms. This interventional study aimed to investigate whether RYGB modulates oxidative stress, inflammation and mitochondrial dynamics in the leukocytes of 47 obese women at one year follow-up. We evaluated biochemical parameters and serum inflammatory cytokines -TNFα, IL6 and IL1β- to assess systemic status. Total superoxide production -dHe-, mitochondrial membrane potential -TMRM-, leucocyte protein expression of inflammation mediators -MCP1 and NF-kB-, antioxidant defence -GPX1-, mitochondrial regulation—PGC1α, TFAM, OXPHOS and MIEAP- and dynamics -MFN2, MNF1, OPA1, FIS1 and p-DRP1- were also determined. After RYGB, a significant reduction in superoxide and mitochondrial membrane potential was evident, while GPX1 content was significantly increased. Likewise, a marked upregulation of the transcription factors PGC1α and TFAM, complexes of the oxidative phosphorylation chain (I–V) and MIEAP and MFN1 was observed. We conclude that women undergoing RYGB benefit from an amelioration of their prooxidant and inflammatory status and an improvement in mitochondrial dynamics of their leukocytes, which is likely to have a positive effect on clinical outcome.
Collapse
Affiliation(s)
- Zaida Abad-Jiménez
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Teresa Vezza
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Sandra López-Domènech
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Meylin Fernández-Reyes
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Francisco Canet
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Carlos Morillas
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Segundo Ángel Gómez-Abril
- Department of General and Digestive System Surgery, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain;
- Department of Surgery, Faculty of Medicine and Dentistry, University of Valencia, Av. Blasco Ibáñez 13, 46010 Valencia, Spain
| | - Celia Bañuls
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Víctor M. Víctor
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
- CIBERehd-Department of Pharmacology, University of Valencia, Av. Blasco Ibáñez 13, 46010 Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Correspondence: (V.M.V.); (M.R.); Tel.: +34-96-318-91-32 (M.R.)
| | - Milagros Rocha
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
- CIBERehd-Department of Pharmacology, University of Valencia, Av. Blasco Ibáñez 13, 46010 Valencia, Spain
- Correspondence: (V.M.V.); (M.R.); Tel.: +34-96-318-91-32 (M.R.)
| |
Collapse
|