1
|
Pandya K, Roul K, Tripathi A, Belemkar S, Sinha A, Erol M, Kumar D. Alzheimer's Disease: A Review of Molecular Mechanisms and Therapeutic Implications by Targeting Sirtuins, Caspases, and GSK-3. ACS Chem Neurosci 2025. [PMID: 40489778 DOI: 10.1021/acschemneuro.5c00207] [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: 06/11/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with a significant impact on global public health. The primary hallmarks of the disease included amyloid-beta peptide (Aβ) deposition, neurofibrillary tangles (NFT), and synaptic loss. Sirtuins, a group of NAD+-dependent deacetylase enzymes, are key regulators of AD pathogenesis. SIRT1, a member of sirtuins, has been identified to possess neuroprotective properties. Thus, its promising enhancers are included. Further, SIRT2 promising inhibitors are reviewed for therapeutic efficacy. The extrinsic and intrinsic apoptotic pathways of caspases are mediated by CD95 and DNA damage. The promising inhibitors Q-VD-OPh and minocycline are found to be specific for caspase-7 and caspase-3, respectively. Primarily, glycogen synthase kinase-3β (GSK-3β) is found to be involved in the generation of phosphorylated tau. The promising GSK-3 inhibitor included the COB-187 (IC50 = 370 nM) and maleimide-derivative (compound 33, IC50 = 0.09 μM). This review highlights the molecular mechanisms of sirtuin, caspase, and GSK-3 in the pathophysiology of AD. Further, promising modulators specific to these targets are described.
Collapse
Affiliation(s)
- Kalpana Pandya
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, V.L. Mehta Road, Vile Parle (West), Mumbai, Maharashtra 400056, India
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management, SVKM's NMIMS University, Mukesh Patel Technology Park, Shirpur 425405, India
| | - Krishnashish Roul
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management, SVKM's NMIMS University, Mukesh Patel Technology Park, Shirpur 425405, India
| | - Avanish Tripathi
- School of Pharmacy, ITM University Gwalior, Gwalior, Madhya Pradesh 475001, India
| | - Sateesh Belemkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, V.L. Mehta Road, Vile Parle (West), Mumbai, Maharashtra 400056, India
| | - Anshuman Sinha
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois IL 60611, United States
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois 60611, United States
| | - Meryem Erol
- Erciyes University, Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Faculty of Pharmacy, Kayseri 38039, Turkey
| | - Devendra Kumar
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management, SVKM's NMIMS University, Mukesh Patel Technology Park, Shirpur 425405, India
| |
Collapse
|
2
|
Zhang H, Chen W, Zhang X, Han Y, Liu W, Li Y, Wang X, Tian D, Xu D, Wang W, YangLi J, Liu Y, Wang X. Signaling pathways underlying extracellular trap formation induced by Vibrio alginolyticus in Strongylocentrotus intermedius. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025:101547. [PMID: 40425409 DOI: 10.1016/j.cbd.2025.101547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 05/29/2025]
Abstract
Extracellular traps (ETs), comprising a DNA-protein network, are widespread and function as an innate immune defense in many species. Notably, Strongylocentrotus intermedius solely depend on innate immunity for disease resistance. This study investigated the formation and preliminary mechanism of ETs in the coelomocytes of the S. intermedius under the stimulation of bacterium Vibrio alginolyticus. These results revealed that as the concentration of V. alginolyticus increased, the formation of ETs became more significant. Flow cytometry analysis showed that the formation process of ETs was accompanied by changes in mitochondrial indicators, suggesting that mitochondria may be involved in the formation process of V. alginolyticus-induced ETs. Transcriptome analysis indicated that the ETs production by coelomocytes of the S. intermedius was related to glycolysis and ATP synthesis. A total of 2631 differentially expressed genes (DEGs) were screened in this transcriptome. We then screened 34 immune-related DEGs from 16 signaling pathways to construct the PPI network, and defined hub proteins corresponding to genes such as ATP6, ND2, G3PDH, MAPK7 and other related genes. These genes are related to mitochondrial function, glycolytic pathways, and immune pathways. Additionally, the formation of ETs led to alterations in multiple immune regulators, such as TNF, NF-κB, MAPK, PI3K-AKT, and mTOR, implying its role in cellular immunomodulation. Quantitative real-time PCR experiment revealed that the expression changes of some DEGs identified and validated in ET-formation coelomocytes matched transcriptome analysis results. This study provided insights into S. intermedius aquaculture, elucidated marine organism immune mechanisms, and advanced invertebrate innate immunity understanding.
Collapse
Affiliation(s)
- Haoyu Zhang
- School of fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Wei Chen
- Yantai Marine Economic Research Institute, Yantai, Shandong 264003, China
| | - Xiaochen Zhang
- School of fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Yijing Han
- School of fisheries, Ludong University, Yantai, Shandong 264025, China.
| | - Wan Liu
- School of fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Yan Li
- School of fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Xiaona Wang
- School of fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Deyang Tian
- Laizhou LiYang Aquatic Development Co., Ltd., Yantai 261441, China
| | - Dong Xu
- Shandong Blue Ocean Technology Co., Ltd., Yantai, 261413, China
| | - Weizhong Wang
- Shandong Blue Ocean Technology Co., Ltd., Yantai, 261413, China
| | - Junsong YangLi
- School of fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Yaqiong Liu
- School of fisheries, Ludong University, Yantai, Shandong 264025, China
| | - Xiaotong Wang
- School of fisheries, Ludong University, Yantai, Shandong 264025, China.
| |
Collapse
|
3
|
Geng J, Yuan Q, Ni C, Zhang Y, Liu X, Zhu X, Hao X, Liang G, Wang D, Fan H. Ca 2+ transfer via enhancing ER-Mito coupling contributed to BDE-47- induced hippocampal neuronal necroptosis and cognitive dysfunction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118396. [PMID: 40412245 DOI: 10.1016/j.ecoenv.2025.118396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 05/11/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
2,2,4,4-Tetrabromodiphenyl ether (BDE-47), a ubiquitous environmental pollutant, has gained increasing attention due to its high level in biological samples and potential neurotoxicity. Recent studies have indicated that the receptor interacting protein kinase 1 (RIPK1)-mediated necroptosis is implicated in BDE-47 cytotoxicity. However, little is known about the underlying mechanism and whether the necroptosis participates in BDE-47-induced neuronal injury and cognitive impairment. Our results indicated that exposure to BDE-47 triggered RIPK1-dependent neuronal necroptosis in mice hippocampi and HT-22 mouse hippocampal neurons. Necrostain-1 (Nec-1), a specific RIPK1 inhibitor, suppressed the RIPK1/RIPK3/mixed lineage kinase-like domain protein (MLKL) signaling and rescued neuronal survival in BDE-47-treated HT-22 neurons. Mechanically, increased mitochondrial Ca2+ influx precipitated the opening of the mitochondrial permeability transition pore (mPTP), leading to occurrence of hippocampal neuronal necroptosis under BDE-47 stress. BDE-47 exposure induced excessive mitochondria-associated endoplasmic reticulum membranes (MAMs) formation and promoted ER-to-mitochondria Ca2+ transfer, while diminishing ER-mitochondrial contacts by Glucose-regulated protein 75 (Grp75)-deficiency remarkably prevented mitochondria Ca2+ overload and opening of mPTP as well as neuronal necroptosis. Notably, Nec-1 pre-treatment could substantially mitigate neuronal/synaptic damage and cognitive impairment in BDE-47-exposed mice. Collectively, these data suggest that BDE-47 exposure intensified endoplasmic reticulum (ER)-mitochondrial (Mito) contact and facilitated Ca2+ transfer from ER towards mitochondria, resulting in mPTP opening-mediated hippocampal neuronal necroptosis and subsequent cognitive dysfunction. Our study shed new light on the mechanisms underlying BDE-47 neurotoxicity and provided a novel therapeutic strategy through targeting RIPK1 kinase activity.
Collapse
Affiliation(s)
- Junhong Geng
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Quan Yuan
- Henan Province Rongkang Hospital, Luoyang, China
| | - Chaofang Ni
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Yarong Zhang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoli Liu
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoying Zhu
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xueqin Hao
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Gaofeng Liang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Dongmei Wang
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China.
| | - Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
4
|
Olatona OA, Sterben SP, Kansakar SBS, Symes AJ, Liaudanskaya V. Mitochondria: the hidden engines of traumatic brain injury-driven neurodegeneration. Front Cell Neurosci 2025; 19:1570596. [PMID: 40417416 PMCID: PMC12098645 DOI: 10.3389/fncel.2025.1570596] [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: 02/03/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025] Open
Abstract
Mitochondria play a critical role in brain energy metabolism, cellular signaling, and homeostasis, making their dysfunction a key driver of secondary injury progression in traumatic brain injury (TBI). This review explores the relationship between mitochondrial bioenergetics, metabolism, oxidative stress, and neuroinflammation in the post-TBI brain. Mitochondrial dysfunction disrupts adenosine triphosphate (ATP) production, exacerbates calcium dysregulation, and generates reactive oxygen species, triggering a cascade of neuronal damage and neurodegenerative processes. Moreover, damaged mitochondria release damage-associated molecular patterns (DAMPs) such as mitochondrial DNA (mtDNA), Cytochrome C, and ATP, triggering inflammatory pathways that amplify tissue injury. We discuss the metabolic shifts that occur post-TBI, including the transition from oxidative phosphorylation to glycolysis and the consequences of metabolic inflexibility. Potential therapeutic interventions targeting mitochondrial dynamics, bioenergetic support, and inflammation modulation are explored, highlighting emerging strategies such as mitochondrial-targeted antioxidants, metabolic substrate supplementation, and pharmacological regulators of mitochondrial permeability transition pores. Understanding these mechanisms is crucial for developing novel therapeutic approaches to mitigate neurodegeneration and enhance recovery following brain trauma.
Collapse
Affiliation(s)
- Olusola A. Olatona
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
| | - Sydney P. Sterben
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
| | - Sahan B. S. Kansakar
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
| | - Aviva J. Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, MD, United States
| | - Volha Liaudanskaya
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
- Neuroscience Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
5
|
Figueiredo Godoy AC, Frota FF, Araújo LP, Valenti VE, Pereira EDSBM, Detregiachi CRP, Galhardi CM, Caracio FC, Haber RSA, Fornari Laurindo L, Tanaka M, Barbalho SM. Neuroinflammation and Natural Antidepressants: Balancing Fire with Flora. Biomedicines 2025; 13:1129. [PMID: 40426956 PMCID: PMC12108937 DOI: 10.3390/biomedicines13051129] [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: 04/06/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Major depressive disorder (MDD) is a major global health concern that is intimately linked to neuroinflammation, oxidative stress, mitochondrial dysfunction, and complicated metabolic abnormalities. Traditional antidepressants frequently fall short, highlighting the urgent need for new, safer, and more acceptable therapeutic techniques. Phytochemicals, i.e., natural antidepressants derived from plants, are emerging as powerful plant-based therapies capable of targeting many pathogenic pathways at the same time. Summary: This narrative review synthesizes evidence from preclinical and clinical studies on the efficacy of phytochemicals such as curcumin, polyphenols, flavonoids, and alkaloids in lowering depressed symptoms. Consistent data show that these substances have neuroprotective, anti-inflammatory, and antioxidant properties, altering neuroimmune interactions, reducing oxidative damage, and improving mitochondrial resilience. Particularly, polyphenols and flavonoids have great therapeutic potential because of their capacity to penetrate the blood-brain barrier, inhibit cytokine activity, and encourage neuroplasticity mediated by brain-derived neurotrophic factor (BDNF). Despite promising results, the heterogeneity in study designs, phytochemical formulations, and patient demographics highlights the importance of thorough, standardized clinical studies. Conclusions: This review identifies phytochemicals as compelling adjuvant or independent therapies in depression treatment, providing multimodal mechanisms and enhanced tolerability. Additional research into improved dosage, pharmacokinetics, long-term safety, and integrative therapy approaches is essential. Using phytotherapeutics could considerably improve holistic and customized depression care, encouraging new research routes in integrative neuroscience and clinical psychiatry.
Collapse
Affiliation(s)
- Ana Clara Figueiredo Godoy
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.P.A.)
| | - Fernanda Fortes Frota
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.P.A.)
| | - Larissa Parreira Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.P.A.)
| | - Vitor E. Valenti
- Autonomic Nervous System Center, School of Philosophy and Sciences, São Paulo State University, Marília 17525-900, SP, Brazil
| | - Eliana de Souza Bastos Mazuqueli Pereira
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.P.A.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.F.L.)
| | - Claudia Rucco P. Detregiachi
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.P.A.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.F.L.)
| | - Cristiano M. Galhardi
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.P.A.)
| | - Flávia Cristina Caracio
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.F.L.)
- School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, SP, Brazil
| | - Rafael S. A. Haber
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.P.A.)
| | - Lucas Fornari Laurindo
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.F.L.)
| | - Masaru Tanaka
- Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Sandra M. Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.P.A.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil (L.F.L.)
- Research Coordinator at UNIMAR Charity Hospital, Marília 17525-902, SP, Brazil
| |
Collapse
|
6
|
Xu C, Jing W, Liu C, Yuan B, Zhang X, Liu L, Zhang F, Chen P, Liu Q, Wang H, Du X. Cytoplasmic DNA and AIM2 inflammasome in RA: where they come from and where they go? Front Immunol 2024; 15:1343325. [PMID: 39450183 PMCID: PMC11499118 DOI: 10.3389/fimmu.2024.1343325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disease of undetermined etiology characterized by symmetric synovitis with predominantly destructive and multiple joint inflammation. Cytoplasmic DNA sensors that recognize protein molecules that are not themselves or abnormal dsDNA fragments play an integral role in the generation and perpetuation of autoimmune diseases by activating different signaling pathways and triggering innate immune signaling pathways and host defenses. Among them, melanoma deficiency factor 2 (AIM2) recognizes damaged DNA and double-stranded DNA and binds to them to further assemble inflammasome, initiating the innate immune response and participating in the pathophysiological process of rheumatoid arthritis. In this article, we review the research progress on the source of cytoplasmic DNA, the mechanism of assembly and activation of AIM2 inflammasome, and the related roles of other cytoplasmic DNA sensors in rheumatoid arthritis.
Collapse
Affiliation(s)
- Conghui Xu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Zheng's Acupuncture, Affiliated Hospital of Gansu University of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Fengfan Zhang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Qiang Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
7
|
Yapryntseva MA, Zhivotovsky B, Gogvadze V. Permeabilization of the outer mitochondrial membrane: Mechanisms and consequences. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167317. [PMID: 38909847 DOI: 10.1016/j.bbadis.2024.167317] [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: 03/31/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
Permeabilization of the outer mitochondrial membrane is а physiological process that can allow certain molecules to pass through it, such as low molecular weight solutes required for cellular respiration. This process is also important for the development of various modes of cell death. Depending on the severity of this process, cells can die by autophagy, apoptosis, or necrosis/necroptosis. Distinct types of pores can be opened at the outer mitochondrial membrane depending on physiological or pathological stimuli, and different mechanisms can be activated in order to open these pores. In this comprehensive review, all these types of permeabilization, the mechanisms of their activation, and their role in various diseases are discussed.
Collapse
Affiliation(s)
- Maria A Yapryntseva
- Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia; Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Boris Zhivotovsky
- Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia; Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | - Vladimir Gogvadze
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
8
|
Zhang L, Hu Z, Li Z, Lin Y. Crosstalk among mitophagy, pyroptosis, ferroptosis, and necroptosis in central nervous system injuries. Neural Regen Res 2024; 19:1660-1670. [PMID: 38103229 PMCID: PMC10960298 DOI: 10.4103/1673-5374.389361] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/28/2023] [Accepted: 09/24/2023] [Indexed: 12/18/2023] Open
Abstract
Central nervous system injuries have a high rate of resulting in disability and mortality; however, at present, effective treatments are lacking. Programmed cell death, which is a genetically determined form of active and ordered cell death with many types, has recently attracted increasing attention due to its functions in determining the fate of cell survival. A growing number of studies have suggested that programmed cell death is involved in central nervous system injuries and plays an important role in the progression of brain damage. In this review, we provide an overview of the role of programmed cell death in central nervous system injuries, including the pathways involved in mitophagy, pyroptosis, ferroptosis, and necroptosis, and the underlying mechanisms by which mitophagy regulates pyroptosis, ferroptosis, and necroptosis. We also discuss the new direction of therapeutic strategies targeting mitophagy for the treatment of central nervous system injuries, with the aim to determine the connection between programmed cell death and central nervous system injuries and to identify new therapies to modulate programmed cell death following central nervous system injury. In conclusion, based on these properties and effects, interventions targeting programmed cell death could be developed as potential therapeutic agents for central nervous system injury patients.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhigang Hu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhenxing Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixing Lin
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| |
Collapse
|
9
|
Oppedisano F, Nesci S, Spagnoletta A. Mitochondrial sirtuin 3 and role of natural compounds: the effect of post-translational modifications on cellular metabolism. Crit Rev Biochem Mol Biol 2024; 59:199-220. [PMID: 38993040 DOI: 10.1080/10409238.2024.2377094] [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: 07/17/2023] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Sirtuins (SIRTs) are a family of proteins with enzymatic activity. In particular, they are a family of class III NAD+-dependent histone deacetylases and ADP-ribosyltransferases. NAD+-dependent deac(et)ylase activities catalyzed by sirtuin include ac(et)ylation, propionylation, butyrylation, crotonylation, manylation, and succinylation. Specifically, human SIRT3 is a 399 amino acid protein with two functional domains: a large Rossmann folding motif and NAD+ binding, and a small complex helix and zinc-binding motif. SIRT3 is widely expressed in mitochondria-rich tissues and is involved in maintaining mitochondrial integrity, homeostasis, and function. Moreover, SIRT3 regulates related diseases, such as aging, hepatic, kidney, neurodegenerative and cardiovascular disease, metabolic diseases, and cancer development. In particular, one of the most significant and damaging post-translational modifications is irreversible protein oxidation, i.e. carbonylation. This process is induced explicitly by increased ROS production due to mitochondrial dysfunction. SIRT3 is carbonylated by 4-hydroxynonenal at the level of Cys280. The carbonylation induces conformational changes in the active site, resulting in allosteric inhibition of SIRT3 activity and loss of the ability to deacetylate and regulate antioxidant enzyme activity. Phytochemicals and, in particular, polyphenols, thanks to their strong antioxidant activity, are natural compounds with a positive regulatory action on SIRT3 in various pathologies. Indeed, the enzymatic SIRT3 activity is modulated, for example, by different natural polyphenol classes, including resveratrol and the bergamot polyphenolic fraction. Thus, this review aims to elucidate the mechanisms by which phytochemicals can interact with SIRT3, resulting in post-translational modifications that regulate cellular metabolism.
Collapse
Affiliation(s)
- Francesca Oppedisano
- Department of Health Sciences, Institute of Research for Food Safety and Health (IRC-FSH), University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Salvatore Nesci
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-Università di Bologna, Ozzano Emilia, Italy
| | - Anna Spagnoletta
- Laboratory "Regenerative Circular Bioeconomy", ENEA-Trisaia Research Centre, Rotondella, Italy
| |
Collapse
|
10
|
Yang Z, Xie Y, Li M, Chen W, Zhong C, Ju J, Deng Q, Wang H, Cheng T, Zhang L, Du W, Liang H. Ramelteon alleviates myocardial ischemia/reperfusion injury (MIRI) through Sirt3--dependent regulation of cardiomyocyte apoptosis. Biomed Pharmacother 2024; 172:116229. [PMID: 38330708 DOI: 10.1016/j.biopha.2024.116229] [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: 10/03/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024] Open
Abstract
Reperfusion stands as a pivotal intervention for ischemic heart disease. However, the restoration of blood flow to ischemic tissue always lead to further damage, which is known as myocardial ischemia/reperfusion injury (MIRI). Ramelteon is an orally administered drug used to improve sleep quality, which is famous for its high bioadaptability and absence of notable addictive characteristics. However, the specific mechanism by which it improves MIRI is still unclear. Sirtuin-3 (Sirt3), primarily located in mitochondria, is crucial in mitigating many cardiac diseases, including MIRI. Based on the structure of Sirt3, we simulated molecular docking and identified several potential amino acid binding sites between it and ramelteon. Therefore, we propose a hypothesis that ramelteon may exert cardioprotective effects by activating the Sirt3 signaling pathway. Our results showed that the activation levels and expression level of Sirt3 were significantly decreased in MIRI tissue and H2O2 stimulated H9C2 cells, while ramelteon treatment upregulated Sirt3 activity and expression. After treat with 3-TYP, a classic Sirt3 activity inhibitor, we constructed myocardial ischemia/reperfusion surgery in vivo and induced H9C2 cells with H2O2 in vitro. The results showed that the myocardial protection and anti-apoptotic effects of ramelteon were antagonized by 3-TYP, indicating that the activation of Sirt3 is a key mechanism for ramelteon to exert myocardial protection. In summary, our results confirm a novel mechanism by which ramelteon improves MIRI by activating Sirt3 signaling pathway, providing strong evidence for the treatment of MIRI with ramelteon.
Collapse
Affiliation(s)
- Zhenbo Yang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China; The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 511400, China
| | - Yilin Xie
- School of Public Health, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Mengyang Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Wenxian Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Changsheng Zhong
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Jin Ju
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 511400, China
| | - Qin Deng
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Huifang Wang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Ting Cheng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Lei Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Weijie Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research,Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| | - Haihai Liang
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai 519000, Guangdong, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
11
|
Cai Z, Chen F, Wang Y, Wang X, Yang X, Zhang C. Lycopene Maintains Mitochondrial Homeostasis to Counteract the Enterotoxicity of Deoxynivalenol. Antioxidants (Basel) 2023; 12:1958. [PMID: 38001811 PMCID: PMC10669674 DOI: 10.3390/antiox12111958] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
The intestinal tract is a target organ for Deoxynivalenol (DON) absorption and toxicity. Mitochondrial homeostasis imbalance is the gut toxicity mechanism of DON. Lycopene (LYC) has intestinal protective effects and can maintain mitochondrial homeostasis in response to various danger signals. The purpose of this study was to explore the protective effect of LYC on DON-induced IPEC-J2 cells damage. These results showed that DON exposure induced an increase in the levels of malondialdehyde and reactive oxygen species (ROS) in IPEC-J2 cells. DON impaired IPEC-J2 cell barrier function and caused mitochondrial dysfunction by inducing mitochondrial permeability transition pore (MPTP) opening, mitochondrial membrane potential (MMP) reducing, destroying mitochondrial fission factors, mitochondrial fusion factors, and mitophagy factors expression. However, adding LYC can reduce the toxic effects of DON-induced IPEC-J2 cells and decrease cellular oxidative stress, functional damage, mitochondrial dynamics imbalance, and mitophagy processes. In conclusion, LYC maintains mitochondrial homeostasis to counteract the IPEC-J2 cells' toxicity of DON.
Collapse
Affiliation(s)
- Zihui Cai
- College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Park, Zhengdong New District, Zhengzhou 450046, China
| | - Fengjuan Chen
- College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Park, Zhengdong New District, Zhengzhou 450046, China
| | - Youshuang Wang
- College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Park, Zhengdong New District, Zhengzhou 450046, China
| | - Xuebing Wang
- College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Park, Zhengdong New District, Zhengzhou 450046, China
| | - Xu Yang
- College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Park, Zhengdong New District, Zhengzhou 450046, China
| | - Cong Zhang
- College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Park, Zhengdong New District, Zhengzhou 450046, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, China
| |
Collapse
|
12
|
Jain A, Casanova D, Padilla AV, Paniagua Bojorges A, Kotla S, Ko KA, Samanthapudi VSK, Chau K, Nguyen MTH, Wen J, Hernandez Gonzalez SL, Rodgers SP, Olmsted-Davis EA, Hamilton DJ, Reyes-Gibby C, Yeung SCJ, Cooke JP, Herrmann J, Chini EN, Xu X, Yusuf SW, Yoshimoto M, Lorenzi PL, Hobbs B, Krishnan S, Koutroumpakis E, Palaskas NL, Wang G, Deswal A, Lin SH, Abe JI, Le NT. Premature senescence and cardiovascular disease following cancer treatments: mechanistic insights. Front Cardiovasc Med 2023; 10:1212174. [PMID: 37781317 PMCID: PMC10540075 DOI: 10.3389/fcvm.2023.1212174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/03/2023] [Indexed: 10/03/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality, especially among the aging population. The "response-to-injury" model proposed by Dr. Russell Ross in 1999 emphasizes inflammation as a critical factor in atherosclerosis development, with atherosclerotic plaques forming due to endothelial cell (EC) injury, followed by myeloid cell adhesion and invasion into the blood vessel walls. Recent evidence indicates that cancer and its treatments can lead to long-term complications, including CVD. Cellular senescence, a hallmark of aging, is implicated in CVD pathogenesis, particularly in cancer survivors. However, the precise mechanisms linking premature senescence to CVD in cancer survivors remain poorly understood. This article aims to provide mechanistic insights into this association and propose future directions to better comprehend this complex interplay.
Collapse
Affiliation(s)
- Ashita Jain
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diego Casanova
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Khanh Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Minh T. H. Nguyen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Jake Wen
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Shaefali P. Rodgers
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | | | - Dale J. Hamilton
- Department of Medicine, Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Cielito Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Xiaolei Xu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Momoko Yoshimoto
- Center for Stem Cell & Regenerative Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, Division of VP Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brain Hobbs
- Department of Population Health, The University of Texas at Austin, Austin, TX, United States
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Efstratios Koutroumpakis
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
13
|
Paik S, Song GY, Jo EK. Ginsenosides for therapeutically targeting inflammation through modulation of oxidative stress. Int Immunopharmacol 2023; 121:110461. [PMID: 37331298 DOI: 10.1016/j.intimp.2023.110461] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/20/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
Ginsenosides are steroid glycosides derived from ginseng plants such as Panax ginseng, Panax quinquefolium, and Panax notoginseng. Advances in recent studies have identified numerous physiological functions of each type of ginsenoside, i.e., immunomodulatory, antioxidative, and anti-inflammatory functions, in the context of inflammatory diseases. Accumulating evidence has revealed the molecular mechanisms by which the single or combined ginsenoside(s) exhibit anti-inflammatory effects, although it remains largely unclear. It is well known that excessive production of reactive oxygen species (ROS) is associated with pathological inflammation and cell death in a variety of cells, and that inhibition of ROS generation ameliorates the local and systemic inflammatory responses. The mechanisms by which ginsenosides attenuate inflammation are largely unknown; however, targeting ROS is suggested as one of the crucial mechanisms for the ginsenosides to control the pathological inflammation in the immune and non-immune cells. This review will summarize the latest progress in ginsenoside studies, particularly in the context of antioxidant mechanisms for its anti-inflammatory effects. A better understanding of the distinct types and the combined action of ginsenosides will pave the way for developing potential preventive and therapeutic modalities in treating various inflammation-related diseases.
Collapse
Affiliation(s)
- Seungwha Paik
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, 35015, South Korea; Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, South Korea.
| | - Gyu Yong Song
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, South Korea; College of Pharmacy, Chungnam National University, Daejeon, 34134, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, 35015, South Korea; Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, South Korea; Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, South Korea.
| |
Collapse
|
14
|
Huang Y, Zhou B. Mitochondrial Dysfunction in Cardiac Diseases and Therapeutic Strategies. Biomedicines 2023; 11:biomedicines11051500. [PMID: 37239170 DOI: 10.3390/biomedicines11051500] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Mitochondria are the main site of intracellular synthesis of ATP, which provides energy for various physiological activities of the cell. Cardiomyocytes have a high density of mitochondria and mitochondrial damage is present in a variety of cardiovascular diseases. In this paper, we describe mitochondrial damage in mitochondrial cardiomyopathy, congenital heart disease, coronary heart disease, myocardial ischemia-reperfusion injury, heart failure, and drug-induced cardiotoxicity, in the context of the key roles of mitochondria in cardiac development and homeostasis. Finally, we discuss the main current therapeutic strategies aimed at alleviating mitochondrial impairment-related cardiac dysfunction, including pharmacological strategies, gene therapy, mitochondrial replacement therapy, and mitochondrial transplantation. It is hoped that this will provide new ideas for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yafei Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing 100037, China
| |
Collapse
|
15
|
Mishra Y, Kumar Kaundal R. Role of SIRT3 in mitochondrial biology and its therapeutic implications in neurodegenerative disorders. Drug Discov Today 2023; 28:103583. [PMID: 37028501 DOI: 10.1016/j.drudis.2023.103583] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/19/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023]
Abstract
Sirtuin 3 (SIRT3), a mitochondrial deacetylase expressed preferentially in high-metabolic-demand tissues including the brain, requires NAD+ as a cofactor for catalytic activity. It regulates various processes such as energy homeostasis, redox balance, mitochondrial quality control, mitochondrial unfolded protein response (UPRmt), biogenesis, dynamics and mitophagy by altering protein acetylation status. Reduced SIRT3 expression or activity causes hyperacetylation of hundreds of mitochondrial proteins, which has been linked with neurological abnormalities, neuro-excitotoxicity and neuronal cell death. A body of evidence has suggested, SIRT3 activation as a potential therapeutic modality for age-related brain abnormalities and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yogesh Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)-226002, India
| | - Ravinder Kumar Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)-226002, India.
| |
Collapse
|
16
|
Abo Alrob O, Al-Horani RA, Altaany Z, Nusair MB. Synergistic Beneficial Effects of Resveratrol and Diet on High-Fat Diet-Induced Obesity. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58091301. [PMID: 36143977 PMCID: PMC9503422 DOI: 10.3390/medicina58091301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022]
Abstract
Introduction: Despite decades of research, obesity and its related medical complications remain a major health concern globally. Therefore, novel therapeutic strategies are needed to combat obesity and its numerous debilitating complications. Resveratrol (RES) has a potential therapeutic effect in obesity and diabetes by improving oxidative metabolism and insulin signaling. Background and Objectives: The aim of this study was to investigate the effect of RES treatment on weight loss and glucose and fatty acid metabolism. Methods: Obesity was induced in 24 mice by exposure to a high-fat diet (HFD) for 8 weeks. Mice were randomly assigned to one group of either: group 1: control, non-treated low-fat diet (LFD) for 12 weeks (n = 8), group 2: non-treated high-fat diet (HFD) for 12 weeks (n = 8), group 3: RES-treated HFD (HFD + RES) (n = 8), or group 4: RES-treated and switched to LFD (HFD-LFD + RES) (n = 8). HFD + RES mice were first fed an HFD for 8 weeks followed by 4 weeks of RES. The HFD-LFD + RES group was first fed an HFD for 8 weeks and then treated with RES and switched to an LFD for 4 weeks. Results: After 12 weeks, group 2 mice had significantly higher body weights compared to group 1 (23.71 ± 1.95 vs. 47.83 ± 2.27; p < 0.05). Group 4 had a significant decrease in body weight and improvement in glucose tolerance compared to mice in group 2 (71.3 ± 1.17 vs. 46.1 ± 1.82 and 40.9 ± 1.75, respectively; p < 0.05). Skeletal muscles expression of SIRT1, SIRT3, and PGC1α were induced in group 3 and 4 mice compared to group 2 (p < 0.01), with no changes in AMP-activated protein kinase expression levels. Furthermore, combination of RES and diet ameliorated skeletal muscle intermediate lipid accumulation and significantly improved insulin sensitivity and secretion. Conclusions: The results of this study suggest a synergistic beneficial effect of LFD and RES to lower body weight and enhance glucose and fatty acid metabolism.
Collapse
Affiliation(s)
- Osama Abo Alrob
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Yarmouk University, Irbid 211-63, Jordan or
- Correspondence:
| | - Ramzi A. Al-Horani
- Department of Exercise Science, Yarmouk University, Irbid 211-63, Jordan
| | - Zaid Altaany
- Department of Basic Sciences, Faculty of Medicine, Yarmouk University, Irbid 211-63, Jordan
| | - Mohammad B. Nusair
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Yarmouk University, Irbid 211-63, Jordan or
- Department of Sociobehavioral and Administrative Pharmacy, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| |
Collapse
|
17
|
Metabolic and Cellular Compartments of Acetyl-CoA in the Healthy and Diseased Brain. Int J Mol Sci 2022; 23:ijms231710073. [PMID: 36077475 PMCID: PMC9456256 DOI: 10.3390/ijms231710073] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022] Open
Abstract
The human brain is characterised by the most diverse morphological, metabolic and functional structure among all body tissues. This is due to the existence of diverse neurons secreting various neurotransmitters and mutually modulating their own activity through thousands of pre- and postsynaptic interconnections in each neuron. Astroglial, microglial and oligodendroglial cells and neurons reciprocally regulate the metabolism of key energy substrates, thereby exerting several neuroprotective, neurotoxic and regulatory effects on neuronal viability and neurotransmitter functions. Maintenance of the pool of mitochondrial acetyl-CoA derived from glycolytic glucose metabolism is a key factor for neuronal survival. Thus, acetyl-CoA is regarded as a direct energy precursor through the TCA cycle and respiratory chain, thereby affecting brain cell viability. It is also used for hundreds of acetylation reactions, including N-acetyl aspartate synthesis in neuronal mitochondria, acetylcholine synthesis in cholinergic neurons, as well as divergent acetylations of several proteins, peptides, histones and low-molecular-weight species in all cellular compartments. Therefore, acetyl-CoA should be considered as the central point of metabolism maintaining equilibrium between anabolic and catabolic pathways in the brain. This review presents data supporting this thesis.
Collapse
|
18
|
Ren G, Ma Y, Wang X, Zheng Z, Li G. Aspirin blocks AMPK/SIRT3-mediated glycolysis to inhibit NSCLC cell proliferation. Eur J Pharmacol 2022; 932:175208. [PMID: 35981603 DOI: 10.1016/j.ejphar.2022.175208] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022]
Abstract
Non-small cell lung cancer (NSCLC) has the highest incidence and mortality in the world. Aspirin has been reported to promote apoptosis, inhibit proliferation, stemness, angiogenesis, cancer-associated inflammation and migration in NSCLC. But the effect of aspirin on aerobic glycolysis in NSCLC is less reported. In the present study, we investigated whether aspirin blocked aerobic glycolysis of NSCLC cells to inhibit proliferation. Our results showed that aspirin inhibited viability, PCNA expression, ability of colony formation, dimished extracellular acidification rate (ECAR), oxygen consumption rate (OCR) and production of pyruvic acid and lactic acid, accompanied with reduced mitochondrial membrane potential (MMP), PGC-1α expression and ROS production, indicating mitochondrial dysfunction in NSCLC cells. AMPK and mitochondrial-localized deacetylase sirtuin 3 (SIRT3) were identified as the relevant molecular targets in glycolysis, but mechanism and relationship between AMPK and SIRT3 for aspirin induced glycolysis inhibition remain unknown in cancer cells. The investigation of underlying mechanism indicated that aspirin activated AMPK pathway to inhibit aerobic glycolysis and proliferation by upregulating SIRT3 after application of compound C (CC), an inhibitor of AMPK activity or SIRT3 siRNA. Upon activation of SIRT3, aspirin promoted the release of hexokinase-II (HK-II) from mitochondrial outer membrane to cytosol by deacetylating cyclophilin D (CypD). Consistently, aspirin significantly inhibited the growth of NSCLC xenografts and exhibited antitumor activity probably through AMPK/SIRT3/HK-II pathway in vivo. Collectively, AMPK/SIRT3/HK-II pathway plays a critical role in anticancer effects of aspirin, and our findings might serve as potential target for clinical practice and chemoprevention of aspirin in NSCLC.
Collapse
Affiliation(s)
- Guanghui Ren
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan, China
| | - Yan Ma
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan, China
| | - Xingjie Wang
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan, China
| | - Zhaodi Zheng
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan, China
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan, China.
| |
Collapse
|
19
|
Yu LM, Dong X, Xu YL, Zhou ZJ, Huang YT, Zhao JK, Xu DY, Xue XD, Zhao QS, Liu T, Yin ZT, Jiang H, Wang HS. Icariin attenuates excessive alcohol consumption-induced susceptibility to atrial fibrillation through SIRT3 signaling. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166483. [DOI: 10.1016/j.bbadis.2022.166483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/25/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
|