1
|
Hayat M, Syed RA, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Albassam LAM, Kaleem I, Wang X, Wang R, Bhatti MS, Bashir S. Decoding molecular mechanisms: brain aging and Alzheimer's disease. Neural Regen Res 2025; 20:2279-2299. [PMID: 39104174 PMCID: PMC11759015 DOI: 10.4103/nrr.nrr-d-23-01403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/23/2024] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
The complex morphological, anatomical, physiological, and chemical mechanisms within the aging brain have been the hot topic of research for centuries. The aging process alters the brain structure that affects functions and cognitions, but the worsening of such processes contributes to the pathogenesis of neurodegenerative disorders, such as Alzheimer's disease. Beyond these observable, mild morphological shifts, significant functional modifications in neurotransmission and neuronal activity critically influence the aging brain. Understanding these changes is important for maintaining cognitive health, especially given the increasing prevalence of age-related conditions that affect cognition. This review aims to explore the age-induced changes in brain plasticity and molecular processes, differentiating normal aging from the pathogenesis of Alzheimer's disease, thereby providing insights into predicting the risk of dementia, particularly Alzheimer's disease.
Collapse
Affiliation(s)
- Mahnoor Hayat
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rafay Ali Syed
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad (IIUI), Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Bioengineering, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | | | - Imdad Kaleem
- Department of Biosciences, Commission on Science and Technology for Sustainable Development in the South (COMSATS University), Islamabad, Pakistan
| | - Xueyi Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Ran Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mehwish S. Bhatti
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
2
|
Ge P, Guo S, Wang P, Zhou P, Tang Z, Yang N, Guo R, Xiao Q, Chai X, Zhang Q, Zhu H. Berbamine targets the FKBP12-rapamycin-binding (FRB) domain of the mTOR complex to promote microglial autophagy and ameliorate neuroinflammation in Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156771. [PMID: 40279970 DOI: 10.1016/j.phymed.2025.156771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/25/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Berbamine (BBM), a natural bisbenzylisoquinoline alkaloid, has demonstrated promising effects in ameliorating pathological process and inflammation response in central neuronal system (CNS). Alzheimer's disease (AD), primarily characterized by amyloid-beta (Aβ)-containing extra-cellular plaques and abnormal "autophagy-brake regulation" of neuroinflammation, currently lacks of effective therapeutic options. Therapeutics of BBM on AD is indeed intriguing, and the potential targets and mechanisms are vague yet. PURPOSE This study is designed to elucidate the therapeutic potential of BBM on AD, focusing particularly on its ability to enhance autophagy, induce microglial M2 polarization, and to uncover the underlying molecular mechanisms and implicated targets. METHODS The therapeutic efficacy of BBM was systematically investigated in APP/PS1 mice, with a focus on its potential to enhance autophagy, induce M2 polarization in microglia, and facilitate the clearance of Aβ plaques. Cognitive function was rigorously assessed through a series of behavioral tests, including the Morris Water Maze and Object Location Task. Immunofluorescence was employed to visualize the spatial distribution of inflammatory cytokines and autophagic markers within the brain parenchyma. Quantitative measurements of these cytokines were obtained using enzyme-linked immunosorbent assay (ELISA). Western blotting was utilized to analyze protein profiles associated with autophagy and microglial phenotypes. Additionally, chemo-proteomics and molecular docking techniques were applied to identify the key molecular targets of BBM. RESULTS BBM treatment significantly ameliorated cognitive dysfunction and reduced Aβ plaque deposition in APP/PS1 transgenic mice. Notably, BBM promoted microglial polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, accompanied by attenuation of neuroinflammation. Mechanistically, BBM exerted its effects through inhibition of mTOR signaling via direct interaction with the FKBP12-rapamycin-binding domain, thereby restoring autophagic flux and facilitating M2 microglial polarization. The mTOR activator MHY1485 abrogated the beneficial effects of BBM, highlighting the pivotal role of mTOR inhibition in its mechanism of action. CONCLUSIONS BBM promotes M2 microglial polarization and restores autophagic flux in AD by inhibiting mTOR signaling, representing a novel dual-modulatory mechanism for AD intervention. These findings highlight BBM's ability to target mTOR and intersecting pathways, offering a promising disease-modifying therapeutic approach for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Pingyuan Ge
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, 138 Xianlin Rd., Nanjing 210023, China
| | - Siqi Guo
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, 138 Xianlin Rd., Nanjing 210023, China
| | - Pingping Wang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, 138 Xianlin Rd., Nanjing 210023, China
| | - Peng Zhou
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, 138 Xianlin Rd., Nanjing 210023, China
| | - Zhishu Tang
- Graduate school, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Nianyun Yang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, 138 Xianlin Rd., Nanjing 210023, China
| | - Rui Guo
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qingqing Xiao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, 138 Xianlin Rd., Nanjing 210023, China
| | - Xin Chai
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qichun Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Huaxu Zhu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, 138 Xianlin Rd., Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing 210023, China.
| |
Collapse
|
3
|
Kshirsagar S, Reddy AP, Reddy PH. Beneficial effects of mitophagy enhancers on amyloid beta-induced mitochondrial and synaptic toxicities in Alzheimer's disease. Mitochondrion 2025; 83:102038. [PMID: 40157622 DOI: 10.1016/j.mito.2025.102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/11/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
The purpose of our study is to investigate the beneficial effects of mitophagy enhancers against mutant amyloid precursor protein (APP) and amyloid beta (Aβ) induced mitochondrial and synaptic toxicities in Alzheimer's disease (AD). Research spanning over two decades highlights the critical role of mitochondrial dysfunction and synaptic damage in the pathogenesis of both early-onset and late-onset AD. Emerging evidence suggests impaired clearance of damaged mitochondria is an early pathological event in AD, positioning mitophagy enhancers as potential therapeutic candidates. This study determined the optimal doses of four mitophagy enhancers-Urolithin A (UA), actinonin, tomatidine, and nicotinamide riboside (NR)-using immortalized mouse hippocampal (HT22) neurons. HT22 cells were transfected with mutant APP (mAPP) cDNA and treated with the enhancers. The effects were assessed by evaluating mRNA and protein expression levels of genes involved in mitochondrial dynamics, biogenesis, mitophagy, and synaptic function, alongside cell survival and mitochondrial respiration. Mitochondrial morphology was also examined in treated and untreated mAPP-HT22 cells. Results showed that mAPP-HT22 cells exhibited increased mitochondrial fission, reduced fusion, downregulated synaptic and mitophagy-related genes, diminished cell survival, impaired mitochondrial respiration, and excessively fragmented, shortened mitochondria. Treatment with mitophagy enhancers reversed these deficits, restoring mitochondrial and synaptic health. Enhanced cell survival, upregulation of mitochondrial fusion, synaptic, and mitophagy genes, improved mitochondrial structure, and reduced fragmentation were observed. Notably, UA demonstrated the most robust mitigating effects. These findings underscore the therapeutic potential of mitophagy enhancers, particularly UA, as promising candidates to treat mitochondrial and synaptic dysfunctions in AD.
Collapse
Affiliation(s)
- Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Arubala P Reddy
- Department of Nutrition, Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
4
|
Pradeepkiran JA, Islam MA, Sehar U, Reddy AP, Vijayan M, Reddy PH. Impact of diet and exercise on mitochondrial quality and mitophagy in Alzheimer's disease. Ageing Res Rev 2025; 108:102734. [PMID: 40120948 DOI: 10.1016/j.arr.2025.102734] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/26/2024] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that affects millions of people worldwide. It is characterized by the accumulation of beta-amyloid and phosphorylated tau, synaptic damage, and mitochondrial abnormalities in the brain, leading to the progressive loss of cognitive function and memory. In AD, emerging research suggests that lifestyle factors such as a healthy diet and regular exercise may play a significant role in delaying the onset and progression of the disease. Mitochondria are often referred to as the powerhouse of the cell, as they are responsible for producing the energy to cells, including neurons to maintain cognitive function. Our article elaborates on how mitochondrial quality and function decline with age and AD, leading to an increase in oxidative stress and a decrease in ATP production. Decline in mitochondrial quality can impair cellular functions contributing to the development and progression of disease with the loss of neuronal functions in AD. This article also covered mitophagy, the process by which damaged or dysfunctional mitochondria are selectively removed from the cell to maintain cellular homeostasis. Impaired mitophagy has been implicated in the progression and pathogenesis of AD. We also discussed the impact of impaired mitophagy implicated in AD, as the accumulation of damaged mitochondria can lead to increased oxidative stress. We expounded how dietary interventions and exercise can help to improve mitochondrial quality, and mitochondrial function and enhance mitophagy in AD. A diet rich in antioxidants, polyphenols, and mitochondria-targeted small molecules has been shown to enhance mitochondrial function and protect against oxidative stress, particularly in neurons with aged and mild cognitively impaired subjects and AD patients. Promoting a healthy lifestyle, mainly balanced diet and regular exercise that support mitochondrial health, in an individual can potentially delay the onset and progression of AD. In conclusion, a healthy diet and regular exercise play a crucial role in maintaining mitochondrial quality and mitochondrial function, in turn, enhancing mitophagy and synaptic activities that delay AD in the elderly populations.
Collapse
Affiliation(s)
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
5
|
Skawratananond S, Xiong DX, Zhang C, Tonk S, Pinili A, Delacruz B, Pham P, Smith SC, Navab R, Reddy PH. Mitophagy in Alzheimer's disease and other metabolic disorders: A focus on mitochondrial-targeted therapeutics. Ageing Res Rev 2025; 108:102732. [PMID: 40122398 DOI: 10.1016/j.arr.2025.102732] [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: 11/15/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Mitochondria, as central regulators of cellular processes such as energy production, apoptosis, and metabolic homeostasis, are essential to cellular function and health. The maintenance of mitochondrial integrity, especially through mitophagy-the selective removal of impaired mitochondria-is crucial for cellular homeostasis. Dysregulation of mitochondrial function, dynamics, and biogenesis is linked to neurodegenerative and metabolic diseases, notably Alzheimer's disease (AD), which is increasingly recognized as a metabolic disorder due to its shared pathophysiologic features: insulin resistance, oxidative stress, and chronic inflammation. In this review, we highlight recent advancements in pharmacological interventions, focusing on agents that modulate mitophagy, mitochondrial uncouplers that reduce oxidative phosphorylation, compounds that directly scavenge reactive oxygen species to alleviate oxidative stress, and molecules that ameliorate amyloid beta plaque accumulation and phosphorylated tau pathology. Additionally, we explore dietary and lifestyle interventions-MIND and ketogenic diets, caloric restriction, physical activity, hormone modulation, and stress management-that complement pharmacological approaches and support mitochondrial health. Our review underscores mitochondria's central role in the pathogenesis and potential treatment of neurodegenerative and metabolic diseases, particularly AD. By advocating for an integrated therapeutic model that combines pharmacological and lifestyle interventions, we propose a comprehensive approach aimed at mitigating mitochondrial dysfunction and improving clinical outcomes in these complex, interrelated diseases.
Collapse
Affiliation(s)
- Shadt Skawratananond
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Daniel X Xiong
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| | - Charlie Zhang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Sahil Tonk
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Aljon Pinili
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Brad Delacruz
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Patrick Pham
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Shane C Smith
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Rahul Navab
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, United States; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
6
|
Li Y, Wang T, Li H, Jiang Y, Shen X, Kang N, Guo Z, Zhang R, Lu X, Kang T, Li M, Hou Y, Wu Y. Targeting LKB1-AMPK-SIRT1-induced autophagy and mitophagy pathways improves cerebrovascular homeostasis in APP/PS1 mice. Free Radic Biol Med 2025; 233:400-418. [PMID: 40180019 DOI: 10.1016/j.freeradbiomed.2025.03.045] [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: 01/24/2025] [Revised: 03/14/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common and severe degenerative disorder of the central nervous system in the elderly, profoundly impacting patients' quality of life. However, effective therapeutic agents for AD are still lacking. Bazi Bushen capsule (BZBS) is a traditional Chinese herbal compound with potential neuroprotective effects, yet its underlying mechanisms remain poorly understood. METHODS In this study, we utilized APP/PS1 transgenic mice to assess the therapeutic efficacy of BZBS. Initially, we evaluated the spatial learning and memory of the mice using the Barnes maze. The brain microcirculation was assessed through a small-animal ultrasound system, two-photon in vivo imaging, and micro-computed tomography angiography. Molecular, biochemical, and pathological analyses were conducted on brain tissues. Through network pharmacology, we identified potential intervention pathways and targets for BZBS in the treatment of AD, which we subsequently validated both in vivo and in vitro. Additionally, we employed molecular virtual docking screening and biolayer interferometry to elucidate the direct interactions of ginsenoside Rg5 and ginsenoside Ro in BZBS with AMPK and LKB1 proteins. RESULTS The BZBS intervention significantly enhanced spatial learning and memory in APP/PS1 mice while decreasing Aβ deposition. Furthermore, BZBS protected cerebrovascular homeostasis and mitigated neuroinflammation, as evidenced by decreased blood-brain barrier permeability, increased expression of tight-junction proteins, and restored cerebral blood flow. Mechanistically, ginsenosides Rg5 and Ro in BZBS directly bind to AMPK and LKB1 proteins, activating the LKB1-AMPK-SIRT1 signaling pathway, promoting autophagy and mitochondrial autophagy, and alleviating oxidative stress damage in endothelial cells. CONCLUSIONS BZBS enhances autophagy-related activity, decreases Aβ deposition, and improves endothelial cell homeostasis through the activation of the LKB1-AMPK-SIRT1 signaling pathway, ultimately leading to improved cognitive function in mice with AD. This study highlights the importance of enhancing autophagic activity and maintaining cerebrovascular homeostasis in mitigating cognitive decline in AD, providing evidence and new insights into the application of compound medicines for treating age-related neurological disorders.
Collapse
Affiliation(s)
- Yawen Li
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Tongxing Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Hongrong Li
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China; Hebei Yiling Hospital, Shijiazhuang, 050035, China
| | - Yuning Jiang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiaogang Shen
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Ning Kang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Zhifang Guo
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Runtao Zhang
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Xuan Lu
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Tianyu Kang
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Mengnan Li
- Hebei Medical University, Shijiazhuang, 050017, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
| | - Yunlong Hou
- Hebei Medical University, Shijiazhuang, 050017, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
| | - Yiling Wu
- Hebei Medical University, Shijiazhuang, 050017, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
| |
Collapse
|
7
|
Li X, Wu Z, Si X, Li J, Wu G, Wang M. The role of mitochondrial dysfunction in the pathogenesis of Alzheimer's disease and future strategies for targeted therapy. Eur J Med Res 2025; 30:434. [PMID: 40450332 DOI: 10.1186/s40001-025-02699-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 05/17/2025] [Indexed: 06/03/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline, behavioral impairments, and psychiatric comorbidities. The pathogenesis of AD remains incompletely elucidated, despite advances in dominant hypotheses such as the β-amyloid (Aβ) cascade, tauopathy, cholinergic deficiency, and neuroinflammation mechanisms. However, these hypotheses inadequately explain the multifactorial nature of AD, which exposes limitations in our understanding of its mechanisms. Mitochondrial dysfunction is known to play a pivotal role in AD, and since patients exhibit intracellular mitochondrial dysfunction and structural changes in the brain at an early stage, correcting the imbalance of mitochondrial homeostasis and the cytopathological changes caused by it may be a potential target for early treatment of AD. Mitochondrial structural abnormalities accelerate AD pathogenesis. For instance, structural and functional alterations in the mitochondria-associated endoplasmic reticulum membrane (MAM) can disrupt intracellular Ca2⁺ homeostasis and cholesterol metabolism, consequently promoting Aβ accumulation. In addition, the overaccumulation of Aβ and hyperphosphorylated tau proteins can further damage neurons by disrupting mitochondrial integrity and mitophagy, thereby amplifying pathological aggregation and exacerbating neurodegeneration in AD. Furthermore, Aβ deposition and abnormal tau proteins can disrupt mitochondrial dynamics through dysregulation of fission/fusion proteins, leading to excessive mitochondrial fragmentation and subsequent dysfunction. Additionally, hyperphosphorylated tau proteins can impair mitochondrial transport, resulting in axonal dysfunction in AD. This article reviews the biological significance of mitochondrial structural morphology, dynamics, and mitochondrial DNA (mtDNA) instability in AD pathology, emphasizing mitophagy abnormalities as a critical contributor to AD progression. Additionally, mitochondrial biogenesis and proteostasis are critical for maintaining mitochondrial function and integrity. Impairments in these processes have been implicated in the progression of AD, further highlighting the multifaceted role of mitochondrial dysfunction in neurodegeneration. It further discusses the therapeutic potential of mitochondria-targeted strategies for AD drug development.
Collapse
Affiliation(s)
- Xin Li
- Department of Neurology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Ziyang Wu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xiaying Si
- Department of Psychiatry, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jing Li
- Department of Neurology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Guode Wu
- Department of Neurology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Manxia Wang
- Department of Neurology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Zeng X, Yuan Y, Li Y, Hu Z, Hu S. Deciphering the NLRP3 inflammasome in diabetic encephalopathy: Molecular insights and emerging therapeutic targets. Exp Neurol 2025; 391:115304. [PMID: 40383363 DOI: 10.1016/j.expneurol.2025.115304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 05/01/2025] [Accepted: 05/11/2025] [Indexed: 05/20/2025]
Abstract
Diabetic encephalopathy (DE) is a neurological complication characterized by neuroinflammation, cognitive impairment, and memory decline, with its pathogenesis closely linked to the activation of the NLRP3 inflammasome. As a central regulator of the innate immune system, the NLRP3 inflammasome plays a pivotal role in DE progression by mediating neuroinflammation, pyroptosis, mitochondrial dysfunction, oxidative stress, endoplasmic reticulum (ER) stress, and microglial polarization. This review systematically explores the molecular mechanisms by which the NLRP3 inflammasome contributes to DE, focusing on its role in neuroinflammatory cascades and neuronal damage, as well as the diabetes-associated physiological changes that exacerbate DE pathogenesis. Furthermore, we summarize emerging therapeutic strategies targeting the NLRP3 inflammasome, including small-molecule inhibitors and bioactive compounds derived from traditional herbal medicine, highlighting their potential for DE treatment. These findings not only advance our understanding of DE but also provide a foundation for developing NLRP3-targeted pharmacological interventions.
Collapse
Affiliation(s)
- Xinyi Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The First Clinical Medical College of Nanchang University, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yi Yuan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; School of Huankui Academy, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yujia Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Shan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
9
|
Shi D, Guo X, Ning Z, Zhang Y, Liu F, Liu M, Wei Y. Review of FUNDC1-mediated mitochondrial autophagy in Alzheimer's disease. Front Aging Neurosci 2025; 17:1544241. [PMID: 40421101 PMCID: PMC12104256 DOI: 10.3389/fnagi.2025.1544241] [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: 12/12/2024] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Mitochondrial autophagy is a critical quality control mechanism that eliminates dysfunctional mitochondria to maintain cellular homeostasis. Among receptor-dependent mitophagy pathways, FUN14 domain-containing 1 (FUNDC1)-a mitochondrial outer membrane protein harboring an LC3-interacting region (LIR)-plays a central role by directly binding to LC3 under stress conditions, thereby initiating autophagosome encapsulation of damaged organelles. Emerging evidence implicates FUNDC1 dysregulation in neurodegenerative diseases, particularly Alzheimer's disease (AD), where defective mitophagy exacerbates hallmark pathologies including Aβ plaque deposition and hyperphosphorylated Tau-driven neurofibrillary tangles. Despite advances, the molecular interplay between FUNDC1 phosphorylation states (e.g., Ser13/Ser17/Tyr18) and AD progression remains poorly defined. This review systematically examines FUNDC1's dual regulatory role in mitophagy, its mechanistic links to Aβ and Tau pathologies, and the therapeutic potential of targeting FUNDC1-associated kinases (e.g., ULK1, CK2) or downstream effectors (e.g., DRP1, OPA1) to counteract mitochondrial dysfunction in AD. By synthesizing recent preclinical and clinical findings, we aim to bridge the gap between FUNDC1 biology and AD therapeutics, highlighting actionable nodes for drug development.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yun Wei
- *Correspondence: Meixia Liu, ; Yun Wei,
| |
Collapse
|
10
|
Chauhan P, Begum MY, Narapureddy BR, Gupta S, Wadhwa K, Singh G, Kumawat R, Sharma N, Ballal S, Jha SK, Abomughaid MM, B D, Ojha S, Jha NK. Unveiling the Involvement of Herpes Simplex Virus-1 in Alzheimer's Disease: Possible Mechanisms and Therapeutic Implications. Mol Neurobiol 2025; 62:5850-5874. [PMID: 39648189 DOI: 10.1007/s12035-024-04535-4] [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: 06/27/2023] [Accepted: 10/01/2024] [Indexed: 12/10/2024]
Abstract
Viruses pose a significant challenge and threat to human health, as demonstrated by the current COVID-19 pandemic. Neurodegeneration, particularly in the case of Alzheimer's disease (AD), is significantly influenced by viral infections. AD is a neurodegenerative disease that affects people of all ages and poses a significant threat to millions of individuals worldwide. The precise mechanism behind its development is not yet fully understood; however, the emergence and advancement of AD can be hastened by various environmental factors, such as bacterial and viral infections. There has been a longstanding suspicion that the herpes simplex virus-1 (HSV-1) may have a role to play in the development or advancement of AD. Reactivation of HSV-1 could potentially lead to damage to neurons, either by direct means or indirectly by triggering inflammation. This article provides an overview of the connection between HSV-1 infections and immune cells (astrocytes, microglia, and oligodendrocytes) in the progression of AD. It summarizes recent scientific research on how HSV-1 affects neurons, which could potentially shed light on the clinical features and treatment options for AD. In addition, the paper has explored the impact of HSV-1 on neurons and its role in various aspects of AD, such as Aβ secretion, tau hyperphosphorylation, metabolic dysregulation, oxidative damage, apoptosis, and autophagy. It is believed that the immune response triggered by HSV-1 reactivation plays a role in the development of neurodegeneration in AD. Despite the lack of a cure for AD, researchers have made significant efforts to study the clinical and pathological aspects of the disease, identify biomarkers, and gain insight into its underlying causes. The goal is to achieve early diagnosis and develop treatments that can modify the progression of the disease. The current article discusses the most promising therapy for combating the viral impacts, which provides additional evidence for the frequent reactivations of latent HSV-1 in the AD brain. However, further research is still required to establish the molecular and cellular mechanisms underlying the development of AD through the reactivation of HSV-1. This could potentially lead to new insights in drug development aimed at preventing HSV-1 reactivation and the subsequent development and progression of AD.
Collapse
Affiliation(s)
- Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India.
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajsthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Suhas Ballal
- Departmant of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Dheepak B
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences & Technology, Galgotias University, Greater Noida, India.
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
11
|
Meng S, Lu Y, Hu J, Luo B, Sun X, Wang X, Jiang Q. Tooth Loss Leads to Cognitive Impairment and Mitochondrial Disturbance in Wistar Rats. Int Dent J 2025; 75:100818. [PMID: 40311189 PMCID: PMC12084507 DOI: 10.1016/j.identj.2025.03.027] [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: 01/13/2025] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND The link between tooth loss and cognitive impairment has become increasingly significant. Recent findings suggest that mitochondrial alteration in hippocampal neurons may mediate this relationship. OBJECTIVE This study aimed to explore the mediating role of mitochondria in the relationship between tooth loss and cognitive function in Wistar rats. METHOD Male Wistar rats (n = 20, 12 weeks old) were randomly divided into tooth extraction (TE) and sham groups. The model was established through upper molar extraction and sham operation respectively. Cognitive evaluations were performed using Morris water maze (MWM) test 8 weeks after the model establishment. Hippocampal neuron morphology was observed. Mitochondrial function was evaluated by ATP level and mitochondrial membrane potential (MMP). Mitophagy assessment involved conducting immunohistochemical and immunofluorescent staining of PTEN-induced kinase 1 (PINK1), Parkin (E3 ubiquitin ligase), translocase of outer mitochondrial membrane 20 (TOMM20), and microtubule-associated protein 1A/1B-light chain 3 (LC3). Additionally, mitophagy protein alterations were analyzed using western blotting. RESULTS Memory impairment in the TE group was obvious 8 weeks after model establishment. Substantial hippocampal mitochondrial dysfunction was observed in the TE group, evidenced by notably decreased ATP production, decreased MMP level, and abnormal mitochondrial morphology in the hippocampus. Diminished mitophagy was detected by immunofluorescent staining, and further confirmed by immunostaining and western blotting, indicating diminished mitophagy marker levels in PINK1 and Parkin, along with decreased LC3II/I ratios and elevated Sequestosome-1 (SQSTM1/P62) levels, highlighting hippocampal mitophagy deficiency following tooth loss. CONCLUSIONS Tooth loss leads to mitochondrial disturbance and inhibits PINK1/Parkin-mediated mitophagy in hippocampal neurons, inducing cognitive impairment. CLINICAL RELEVANCE This study reveals mitochondria may mediate the effect of tooth loss on cognitive function, offering a theoretical basis for the prevention of oral health-associated cognitive decline.
Collapse
Affiliation(s)
- Shixiang Meng
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Yunping Lu
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Jiangqi Hu
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Bin Luo
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Xu Sun
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Xiaoyu Wang
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Qingsong Jiang
- Department of Prosthodontics, Beijing Stomatology Hospital & School of Stomatology, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Wang SY, Chen YS, Jin BY, Bilal A. The cGAS-STING pathway in atherosclerosis. Front Cardiovasc Med 2025; 12:1550930. [PMID: 40351606 PMCID: PMC12062000 DOI: 10.3389/fcvm.2025.1550930] [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: 12/24/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Atherosclerosis (AS), a chronic inflammatory disease, remains a leading contributor to cardiovascular morbidity and mortality. Recent studies highlight the critical role of the cGAS-STING pathway-a key innate immune signaling cascade-in driving AS progression. This pathway is activated by cytoplasmic DNA from damaged cells, thereby triggering inflammation and accelerating plaque formation. While risk factors such as aging, obesity, smoking, hypertension, and diabetes are known to exacerbate AS, emerging evidence suggests that these factors may also enhance cGAS-STING pathway, which amplifies inflammatory responses. Targeting this pathway offers a promising therapeutic strategy to reduce the burden of cardiovascular diseases (CVD). In this review, we summarize the mechanisms of the cGAS-STING pathway, explore its role in AS, and evaluate potential inhibitors as future therapeutic candidates. By integrating current knowledge, we aim to provide insights for developing novel treatments to mitigate AS and CVD burden.
Collapse
Affiliation(s)
- Si-yu Wang
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Yu-shan Chen
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Bo-yuan Jin
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Ahmad Bilal
- Department of Cardiology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
- The First Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Zhu S, Tang M, Chen J, Li S, Xue R. Mitophagy Protects Against Cisplatin-Induced Injury in Granulosa Cells. TOXICS 2025; 13:332. [PMID: 40423411 DOI: 10.3390/toxics13050332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025]
Abstract
Cisplatin, a widely used chemotherapeutic agent, is known to induce premature ovarian insufficiency (POI) and infertility in women of reproductive age. Among the contributing factors, cisplatin-induced apoptosis of ovarian granulosa cells is considered a primary driver of ovarian dysfunction; however, the underlying mechanisms remain incompletely understood. In this study, we investigated the cytotoxicity of cisplatin on the granulosa cell line KGN in vitro and explored the associated mechanisms. Our results demonstrate that cisplatin induces KGN cell apoptosis in a dose-dependent manner and impairs mitochondrial function, as evidenced by excessive ROS production, membrane potential collapse, and reduced ATP synthesis. Mitophagy, a key cellular self-protection mechanism that selectively removes damaged mitochondria, was activated following cisplatin treatment, mitigating its detrimental effects on KGN cells. Activation of mitophagy with urolithin A (UA) ameliorated cisplatin-induced mitochondrial dysfunction and apoptosis, whereas inhibition of mitophagy with cyclosporine A (CsA) exacerbated these effects. Furthermore, pretreatment with the clinical drug melatonin significantly enhanced mitophagy, effectively attenuating cisplatin-induced apoptosis in KGN cells. This study proposes a novel therapeutic strategy for patients undergoing tumor chemotherapy, aiming to preserve treatment efficacy while reducing the adverse effects of chemotherapeutic agents on ovarian function, thereby improving patients' quality of life.
Collapse
Affiliation(s)
- Sihui Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230088, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei 230032, China
| | - Mingge Tang
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei 230032, China
| | - Jiahua Chen
- Anhui Province Key Laboratory of Reproductive Disorders and Obstetrics and Gynaecology Diseases, Hefei 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei 230032, China
| | - Shuhang Li
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230031, China
| | - Rufeng Xue
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230088, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei 230032, China
| |
Collapse
|
14
|
Basak B, Holzbaur ELF. Mitophagy in Neurons: Mechanisms Regulating Mitochondrial Turnover and Neuronal Homeostasis. J Mol Biol 2025:169161. [PMID: 40268233 DOI: 10.1016/j.jmb.2025.169161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
Mitochondrial quality control is instrumental in regulating neuronal health and survival. The receptor-mediated clearance of damaged mitochondria by autophagy, known as mitophagy, plays a key role in controlling mitochondrial homeostasis. Mutations in genes that regulate mitophagy are causative for familial forms of neurological disorders including Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS). PINK1/Parkin-dependent mitophagy is the best studied mitophagy pathway, while more recent work has brought to light additional mitochondrial quality control mechanisms that operate either in parallel to or independent of PINK1/Parkin mitophagy. Here, we discuss our current understanding of mitophagy mechanisms operating in neurons to govern mitochondrial homeostasis. We also summarize progress in our understanding of the links between mitophagic dysfunction and neurodegeneration, and highlight the potential for therapeutic interventions to maintain mitochondrial health and neuronal function.
Collapse
Affiliation(s)
- Bishal Basak
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
15
|
Yan W, Saqirile, Li K, Li K, Wang C. The Role of N6-Methyladenosine in Mitochondrial Dysfunction and Pathology. Int J Mol Sci 2025; 26:3624. [PMID: 40332101 PMCID: PMC12026702 DOI: 10.3390/ijms26083624] [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: 02/28/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Mitochondria are indispensable in cells and play crucial roles in maintaining cellular homeostasis, energy production, and regulating cell death. Mitochondrial dysfunction has various manifestations, causing different diseases by affecting the diverse functions of mitochondria in the body. Previous studies have mainly focused on mitochondrial-related diseases caused by nuclear gene mutations or mitochondrial gene mutations, or mitochondrial dysfunction resulting from epigenetic regulation, such as DNA and histone modification. In recent years, as a popular research area, m6A has been involved in a variety of important processes under physiological and pathological conditions. However, there are few summaries on how RNA methylation, especially m6A RNA methylation, affects mitochondrial function. Additionally, the role of m6A in pathology through influencing mitochondrial function may provide us with a new perspective on disease treatment. In this review, we summarize several manifestations of mitochondrial dysfunction and compile examples from recent years of how m6A affects mitochondrial function and its role in some diseases.
Collapse
Affiliation(s)
| | | | | | | | - Changshan Wang
- School of Life Science, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (S.); (K.L.); (K.L.)
| |
Collapse
|
16
|
Zuo X. Mitochondrial Imbalance in Down Syndrome: A Driver of Accelerated Brain Aging? Aging Dis 2025:AD.2025.0189. [PMID: 40249934 DOI: 10.14336/ad.2025.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/06/2025] [Indexed: 04/20/2025] Open
Abstract
Down syndrome (DS), caused by trisomy of chromosome 21 (HSA21), is a complex condition associated with neurodevelopmental impairments and accelerated brain aging, often culminating in early-onset Alzheimer's disease (AD). Central to this accelerated aging is mitochondrial imbalance, characterized by disrupted energy metabolism, increased oxidative stress, impaired dynamics, and defective quality control mechanisms like mitophagy. These abnormalities exacerbate neuronal vulnerability, driving cognitive decline and neurodegeneration. This review examines the genetic and biochemical underpinnings of mitochondrial dysfunction in DS, with a focus on the role of HSA21-encoded genes. We also highlight how mitochondrial dysfunction, amplified by oxidative stress and HSA21 gene dosage effects, converges with cellular senescence and neuroinflammation to accelerate Alzheimer-like pathology and brain aging in DS. Finally, we discuss emerging therapeutic strategies targeting mitochondrial pathways, which hold promise for mitigating neurodegenerative phenotypes and improving outcomes in DS.
Collapse
|
17
|
Lin M, Tang K, Zheng W, Zheng S, Hu K. Curcumin delivery system based on a chitosan-liposome encapsulated zeolitic imidazolate framework-8: a potential treatment antioxidant and antibacterial treatment after phacoemulsification. Biomed Mater 2025; 20:035013. [PMID: 40081008 DOI: 10.1088/1748-605x/adc05c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/13/2025] [Indexed: 03/15/2025]
Abstract
Curcumin is a natural polyphenol extracted from plants that can interact with various molecular targets, including antioxidant, antibacterial, anticancer, and anti-aging activities. Due to its variety of pharmacological activities and large margin pf safety, curcumin has been used in the prevention and treatment of various diseases, such as Alzheimer's, heart, and rheumatic immune diseases. To develop curcumin eye drops that can be used as antioxidant and antibacterial agents after phacoemulsification, we have designed a nano-based drug delivery system to improve curcumin bioavailability and duration of action. We successfully prepared zeolitic imidazolate framework-8 (ZIF-8) coated with chitosan-liposome (Cur@ZIF-8/CS-Lip) for curcumin delivery. It can release curcumin for over 20 hin vitroand exhibits excellent biosafety, antioxidant, and antibacterial activities. Therefore, we hypothesized that Cur@ZIF-8/CS-Lip could reduce the incidence of oxidative stress and infection after cataract surgery.
Collapse
Affiliation(s)
- Meiting Lin
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Kunyuan Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Wendi Zheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Shijie Zheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| |
Collapse
|
18
|
MacMullen C, Sharma N, Davis RL. Mitochondrial dynamics and bioenergetics in Alzheimer's induced pluripotent stem cell-derived neurons. Brain 2025; 148:1405-1420. [PMID: 39513728 DOI: 10.1093/brain/awae364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Abstract
Mitochondrial dysfunction is a hallmark of Alzheimer's disease, but the scope and severity of these specific deficits across forms of Alzheimer's disease are not well characterized. We designed a high-throughput longitudinal phenotypic assay to track mitochondrial dynamics and bioenergetics in glutamatergic induced pluripotent stem cell (iPSC)-derived human neurons possessing mutations in presenilin 1 (PSEN1), presenilin 2 (PSEN2) and the amyloid beta precursor protein (APP). Each gene set was composed of iPSC-derived neurons from an Alzheimer's disease patient in addition to two to three engineered mutations with appropriate isogenic and age-matched controls. These iPSC-derived neurons were imaged every other day, beginning at 10 days in vitro, to assess how mitochondrial length and content change over a 10 day time course using a mitochondrially targeted reporter. A second cytosolic reporter allowed for visualization of neurites. Bioenergetics assays, focusing on mitochondrial respiration and individual electron transport chain complexes, were also surveyed over this time course. Mutations in all three genes altered mitochondrial function measured by basal, ATP-linked and maximal oxygen consumption rates and by spare respiratory capacity, with PSEN1/PSEN2 alleles being more severe than APP mutations. Electron flow through Complexes I-IV was decreased in PSEN1/PSEN2 mutations but, in contrast, APP alleles had only modest impairments of complexes I and II. We measured aspects of mitochondrial dynamics, including fragmentation and neurite degeneration, both of which were dramatic in PSEN1/PSEN2 alleles, but essentially absent in APP alleles. The marked differences in mitochondrial pathology might occur from the distinct ways in which amyloids are processed into amyloid beta peptides and might be correlated with the disease severity.
Collapse
Affiliation(s)
- Courtney MacMullen
- Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Neelam Sharma
- Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Ronald L Davis
- Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| |
Collapse
|
19
|
Zhou Q, Wang W, Deng C. Advancements in Proteolysis Targeting Chimeras for Targeted Therapeutic Strategies in Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04838-0. [PMID: 40133753 DOI: 10.1007/s12035-025-04838-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/09/2025] [Indexed: 03/27/2025]
Abstract
The presence of hyperphosphorylated Tau proteins, which mislocalize and form neurofibrillary tangles, and the accumulation of amyloid-β plaques are hallmark features of Alzheimer's disease (AD). These toxic protein aggregates contribute to synaptic impairment and neuronal dysfunction, underscoring the need for strategies aimed at effectively clearing or reducing these aggregates in the treatment of AD. In recent years, proteolysis targeting chimera (PROTAC) technology has emerged as a promising approach for selectively degrading dysfunctional proteins rather than merely inhibiting their function. This approach holds great potential for developing more effective interventions that could slow AD progression and improve patient outcomes. In this review, we first examine the pathological mechanisms underlying AD, focusing on abnormal protein degradation and accumulation. We then explore the evolution of PROTAC technology, its mechanisms of action, and the current status of drug development. Finally, we discuss the latest findings regarding the application of PROTACs in AD therapy, highlighting the potential benefits and limitations of this technology. Although promising, further clinical research is necessary to fully assess the safety and efficacy of PROTAC-based therapies for AD treatment.
Collapse
Affiliation(s)
- Qiuzhi Zhou
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weixia Wang
- School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
20
|
Elazab ST, Hsu WH. α-Bisabolol alleviates doxorubicin-induced cognitive dysfunction in rats via enhancing the hippocampal BDNF/TrKB signaling and inhibiting neuroinflammation. Front Pharmacol 2025; 16:1549009. [PMID: 40124785 PMCID: PMC11925949 DOI: 10.3389/fphar.2025.1549009] [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: 12/20/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Chemofog is a serious sequela commonly manifested among cancer patients receiving doxorubicin (DOX) chemotherapy. Our goal was to explore the abrogative action of α-Bisabolol (BISA), a phytochemical sesquiterpene, against DOX-induced cognitive deficit. Rats were allocated into 5 groups: Group I: control; Group II received BISA orally (100 mg/kg/day for 4 weeks); Group III received DOX (2 mg/kg/week/i.p.) for 4 weeks; Groups IV and V were administered BISA orally at 50 and 100 mg/kg, respectively plus DOX, i. p. Results: 1) BISA attenuated DOX-induced chemofog as shown in memory-related behavioral tests. 2) BISA restored the hippocampal histological structure and redox homeostasis via diminishing MDA content and upregulating Nrf2 and HO-1 genes. 3) BISA mitigated DOX-induced neuroinflammation through reducing NF-kB, TNF-α, IL-6, IL-1β, and GFAP expressions. 4) BISA repressed the hippocampal apoptosis via downregulating Bax gene and upregulating Bcl-2 gene. 5) BISA enhanced the synaptic plasticity by activating the BDNF/TrKB signaling and increasing the levels of neurotransmitters that enhance memory, i.e., ACh, 5-HT, and DA. BISA at 100 mg/kg/day exerted a better neuroprotection than BISA at 50 mg/kg/day. Thus, BISA may protect cancer patients from cognitive disorders caused by DOX.
Collapse
Affiliation(s)
- Sara T. Elazab
- Department of Pharmacology, Facultyof Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Walter H. Hsu
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
21
|
Zhu CZ, Li GZ, Lyu HF, Lu YY, Li Y, Zhang XN. Modulation of autophagy by melatonin and its receptors: implications in brain disorders. Acta Pharmacol Sin 2025; 46:525-538. [PMID: 39448859 PMCID: PMC11845611 DOI: 10.1038/s41401-024-01398-2] [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: 06/27/2024] [Accepted: 09/17/2024] [Indexed: 10/26/2024]
Abstract
Autophagy plays a crucial role in maintaining neuronal homeostasis and function, and its disruption is linked to various brain diseases. Melatonin, an endogenous hormone that primarily acts through MT1 and MT2 receptors, regulates autophagy via multiple pathways. Growing evidence indicates that melatonin's ability to modulate autophagy provides therapeutic and preventive benefits in brain disorders, including neurodegenerative and affective diseases. In this review, we summarize the key mechanisms by which melatonin affects autophagy and explore its therapeutic potential in the treatment of brain disorders.
Collapse
Affiliation(s)
- Chen-Ze Zhu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Gui-Zhi Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, China
| | - Hai-Feng Lyu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yang-Yang Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Yue Li
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China
| | - Xiang-Nan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, 310058, China.
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China.
| |
Collapse
|
22
|
Yang Q, Chen T, Li S, Yang C, Zheng X, Mao S, Liu N, Mo S, Li D, Yang M, Lu Z, Tang L, Huang X, Liu X, Jian C, Yin Y, Shang J. Inhibition of autophagy attenuates cognitive decline and mitochondrial dysfunction in an Alzheimer's disease mouse model with chronic cerebral hypoperfusion. Brain Res 2025; 1850:149416. [PMID: 39710054 DOI: 10.1016/j.brainres.2024.149416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
This study aimed to investigate the impact of chronic cerebral hypoperfusion (CCH) on cognitive function, amyloid-β (Aβ) deposition, cellular autophagy, and mitochondrial dynamics in an Alzheimer's disease (AD) mouse model, and to evaluate the intervention effects of autophagy modulation on these outcomes. Utilizing the APP/PS1 mouse model combined with CCH, we assessed cognitive function, Aβ deposition, and the expression levels of relevant proteins through behavioral tests and immunohistochemical analysis. Our findings revealed pronounced cognitive deficits and increased Aβ deposition in the AD + CCH group mice, along with upregulation of mitochondrial fission proteins (Drp1, Fis1) and downregulation of mitochondrial fusion proteins (Opa1, Mfn1), indicating a shift towards mitochondrial fission and promoting cell apoptosis. Additionally, alterations were observed in the expression levels of cellular autophagy-related proteins (LC3-II, P62), which were reversed by treatment with autophagic inhibitor 3-methyladenine (3-MA). Furthermore, the expression of mitochondrial autophagy-related proteins PINK1 and Parkin was affected, with 3-MA alleviating this effect. In summary, our study elucidates the complex interplay among cognitive decline, increased Aβ deposition, and mitochondrial dysfunction in the AD + CCH model, and suggests that modulating autophagy could be a potential therapeutic strategy for treating the AD + CCH model.
Collapse
Affiliation(s)
- Qin Yang
- Medical School, Jinan University, Guangzhou, Guangdong, China; Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China; Department of Neurology, Baise People's Hospital, Baise, Guangxi, China
| | - Tingting Chen
- School of Basic Medicine, Guangdong Medical University, DongGuang, China
| | - Shaofa Li
- Department of Neurology, Baise People's Hospital, Baise, Guangxi, China
| | - Chengmin Yang
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xingwu Zheng
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Sanying Mao
- Department of Neurology, The First People's Hospital of Jiande, Hangzhou, China
| | - Ning Liu
- School of Basic Medical Sciences, Beihua University, Jilin, China
| | - Shenglong Mo
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Dengxing Li
- Department of Neurology, Baise People's Hospital, Baise, Guangxi, China
| | - Meiling Yang
- Graduate School of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Zhicheng Lu
- Graduate School of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Lina Tang
- Graduate School of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xiaorui Huang
- Department of Psychiatry and Psychology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xia Liu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Chongdong Jian
- Medical School, Jinan University, Guangzhou, Guangdong, China; Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China; Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Baise, Guangxi, China.
| | - Yixia Yin
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China.
| | - Jingwei Shang
- Department of Neurology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China; Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Baise, Guangxi, China.
| |
Collapse
|
23
|
Jiao B, Ouyang Z, Xiao X, Zhang C, Xu T, Yang Q, Zhu Y, Liu Y, Liu X, Zhou Y, Liao X, Luo S, Tang B, Li Z, Shen L. Development and validation of machine learning models with blood-based digital biomarkers for Alzheimer's disease diagnosis: a multicohort diagnostic study. EClinicalMedicine 2025; 81:103142. [PMID: 40115175 PMCID: PMC11925590 DOI: 10.1016/j.eclinm.2025.103142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/23/2025] Open
Abstract
Background Alzheimer's disease (AD) involves complex alterations in biological pathways, making comprehensive blood biomarkers crucial for accurate and earlier diagnosis. However, the cost-effectiveness and operational complexity of method using blood-based biomarkers significantly limit its availability in clinical practice. Methods We developed low-cost, convenient machine learning-based with digital biomarkers (MLDB) using plasma spectra data to detect AD or mild cognitive impairment (MCI) from healthy controls (HCs) and discriminate AD from different types of neurodegenerative diseases. Retrospective data were gathered for 1324 individuals, including 293 with amyloid beta positive AD, 151 with mild cognitive impairment (MCI), 106 with Lewy body dementia (DLB), 106 with frontotemporal dementia (FTD), 135 with progressive supranuclear palsy (PSP) and 533 healthy controls (HCs) between July 2017 and August 2023. Findings Random forest classifier and feature selection procedures were used to select digital biomarkers. MLDB achieved area under the curves (AUCs) of 0.92 (AD vs. HC, Sensitivity 88.2%, specificity 84.1%), 0.89 (MCI vs. HC, Sensitivity 88.8%, specificity 86.4%), 0.83 (AD vs. DLB, Sensitivity 77.2%, specificity 74.6%), 0.80 (AD vs. FTD, sensitivity 74.2%, specificity 72.4%), and 0.93 (AD vs. PSP, sensitivity 76.1%, specificity 75.7%). Digital biomarkers distinguishing AD from HC were negatively correlated with plasma p-tau217 (r = -0.22, p < 0.05) and glial fibrillary acidic protein (GFAP) (r = -0.09, p < 0.05). Interpretation The ATR-FTIR (Attenuated Total Reflectance-Fourier Transform Infrared) plasma spectra features can identify AD-related pathological changes. These spectral features serve as digital biomarkers, providing valuable support in the early screening and diagnosis of AD. Funding The National Natural Science Foundation of China, STI2030-Major Projects, National Key R&D Program of China, Outstanding Youth Fund of Hunan Provincial Natural Science Foundation, Hunan Health Commission Grant, Science and Technology Major Project of Hunan Province, Hunan Innovative Province Construction Project, Grant of National Clinical Research Center for Geriatric Disorders, Xiangya Hospital and Postdoctoral Fellowship Program of CPSF.
Collapse
Affiliation(s)
- Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Brain Research Center, Central South University, Changsha, China
- FuRong Laboratory, Changsha, China
| | - Ziyu Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Cong Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Tianyan Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yiliang Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xixi Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Yafang Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Brain Research Center, Central South University, Changsha, China
- FuRong Laboratory, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxin Liao
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Brain Research Center, Central South University, Changsha, China
- FuRong Laboratory, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Shilin Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Brain Research Center, Central South University, Changsha, China
- FuRong Laboratory, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Brain Research Center, Central South University, Changsha, China
- FuRong Laboratory, Changsha, China
| | - Zhigang Li
- College of Information Science and Engineering, Northeastern University, Shenyang, China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Brain Research Center, Central South University, Changsha, China
- FuRong Laboratory, Changsha, China
| |
Collapse
|
24
|
Dong Q, Zhu Y, Zhang X, Li L, Yang Y, Liu C, Wen J. Phytochemicals Targeting Mitophagy to Treat Heart Diseases: Retrospective Insights and Prospective Directions. Phytother Res 2025; 39:1592-1614. [PMID: 39912509 DOI: 10.1002/ptr.8448] [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: 11/28/2024] [Revised: 01/07/2025] [Accepted: 01/19/2025] [Indexed: 02/07/2025]
Abstract
Mitophagy is a process by which cells selectively eliminate damaged or dysfunctional mitochondria through the autophagy-lysosome pathway, thereby maintaining mitochondrial quality and cellular homeostasis. This process is closely linked to the onset and progression of various heart diseases. Modern pharmacological research has demonstrated that phytochemicals can regulate mitochondrial homeostasis in cardiomyocytes through multiple mechanisms, influencing mitophagy and protecting cardiomyocytes, which in turn exerts anti-cardiovascular effects. However, the underlying mechanisms of these effects are not yet fully understood. This study summarizes the pharmacological effects and molecular mechanisms of mitophagy in heart diseases, aiming to provide reference for the research and treatment of phytochemicals targeting mitophagy against heart diseases. The results indicated that phytochemicals (such as Berberine, Ginsenoside Rg1, Quercetin, Resveratrol, Baicalein, and so on) can exert preventive and therapeutic effects on heart diseases (such as cardiac toxicity or damage, myocardial ischemia/reperfusion injury, heart failure, heart aging, cardiac hypertrophy, cardiomyopathy, and so on.) via regulating the PINK1/Parkin and FUNDC1-dependent mitophagy pathway. These compounds mainly exert their effects by regulating mitochondrial homeostasis, mitochondrial dynamics, mitochondrial oxidative stress, mitochondrial apoptosis, and mitochondrial energy metabolism. This study provides a reference that phytochemicals have effect on anti-cardiovascular effects by regulating mitophagy. However, further in-depth mechanistic and clinical research are needed in the future.
Collapse
Affiliation(s)
- Qin Dong
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yichan Zhu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Xinghai Zhang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Lu Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yi Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Chuan Liu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Jianxia Wen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| |
Collapse
|
25
|
Liu X, Li T, Tu X, Xu M, Wang J. Mitochondrial fission and fusion in neurodegenerative diseases:Ca 2+ signalling. Mol Cell Neurosci 2025; 132:103992. [PMID: 39863029 DOI: 10.1016/j.mcn.2025.103992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Neurodegenerative diseases (NDs) are a group of disorders characterized by the progressive loss of neuronal structure and function. The pathogenesis is intricate and involves a network of interactions among multiple causes and systems. Mitochondria and Ca2+ signaling have long been considered to play important roles in the development of various NDs. Mitochondrial fission and fusion dynamics are important processes of mitochondrial quality control, ensuring the stability of mitochondrial structure and function. Mitochondrial fission and fusion imbalance and Ca2+ signaling disorders can aggravate the disease progression of NDs. In this review, we explore the relationship between mitochondrial dynamics and Ca2+ signaling in AD, PD, ALS, and HD, focusing on the roles of key regulatory proteins (Drp1, Fis1, Mfn1/2, and Opa1) and the association structures between mitochondria and the endoplasmic reticulum (MERCs/MAMs). We provide a detailed analysis of their involvement in the pathogenesis of these four NDs. By integrating these mechanisms, we aim to clarify their contributions to disease progression and offer insights into the development of therapeutic strategies that target mitochondrial dynamics and Ca2+ signaling. We also examine the progress in drug research targeting these pathways, highlighting their potential as therapeutic targets in the treatment of NDs.
Collapse
Affiliation(s)
- Xuan Liu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Tianjiao Li
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Xinya Tu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Mengying Xu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Jianwu Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| |
Collapse
|
26
|
Hu L, Liu J, Peng J, Li X, Huang Z, Zhang C, Fan S. TREM2 Alleviates Neuroinflammation by Maintaining Cellular Metabolic Homeostasis and Mitophagy Activity During Early Inflammation. Diseases 2025; 13:60. [PMID: 39997067 PMCID: PMC11854088 DOI: 10.3390/diseases13020060] [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: 01/12/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
AIMS Inflammation is a pivotal characteristic of neurodegenerative diseases. The triggering receptor expressed on the myeloid cells 2 (TREM2) gene has previously been shown to suppress inflammation by directly inhibiting inflammation-related pathways. Mitochondrial dysfunction has recently emerged as another critical pathological manifestation of neurodegenerative diseases. Although TREM2 is involved in the regulation of cellular energy metabolism and mitochondrial autophagy, its role in the relationship between inflammation and mitochondrial autophagy remains unclear. METHODS In this study, we generated TREM2-overexpressing BV-2 cells and established a neuroinflammatory model with LPS. We compared these cells with wild-type cells in terms of inflammation, metabolism, autophagy, and mitochondria using methods such as RT-qPCR, Western blotting, immunocytochemistry, transmission electron microscopy, and flow cytometry. RESULTS Microglia overexpressing TREM2 exhibited increased resistance to inflammation. Additionally, these cells inhibited the metabolic reprogramming that occurs early in LPS-induced inflammation, reduced ROS release, mitigated mitochondrial damage, maintained a certain level of autophagic activity, and cleared damaged mitochondria. Consequently, they alleviated the inflammation caused by the mitochondrial barrier. CONCLUSIONS ur results suggest that TREM2 can alleviate inflammation by maintaining cellular metabolic homeostasis and mitochondrial autophagy activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shengtao Fan
- Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming 650108, China; (L.H.); (J.L.); (J.P.); (X.L.); (Z.H.); (C.Z.)
| |
Collapse
|
27
|
Lin L, Li J, Yu Z, He J, Li Y, Jiang J, Xia Y. Nrf2 activator tertiary butylhydroquinone enhances neural stem cell differentiation and implantation in Alzheimer's disease by boosting mitochondrial function. Brain Res 2025; 1849:149341. [PMID: 39566569 DOI: 10.1016/j.brainres.2024.149341] [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: 06/27/2024] [Revised: 09/24/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
AIMS To investigate the effects of Nrf2 agonist tertiary butylhydroquinone (TBHQ)-stimulated neural stem cells (NSCs) transplantation (NSC(TBHQ)) on neuronal damage and cognitive deficits in an AD model and its underlying principles. METHODS BHQ-treated NSCs were examined with or without Aβ1-42 to investigate the effects of TBHQ on the proliferation and differentiation functions. The mitophagy inhibitor Cyclosporine A (CSA) was used to explore the regulation of mitophagy by TBHQ. The no-, ethanol-, and TBHQ-treated NSCs were transplanted into the bilateral hippocampal region of model mice to explore the effects of NSC(TBHQ) on neuronal, cognitive, and mitochondrial functional impairments in mice. RESULTS TBHQ reversed the Aβ1-42-caused inhibition on NSC proliferation and differentiation, as well as on levels of mitochondrial membrane potential, adenosine triphosphate (ATP), and mitochondrial fusion-associated proteins. TBHQ alleviated the Aβ1-42-induced increase in apoptosis, mitochondrial damage, mitochondria-derived reactive oxygen species (mtROS), and mitochondrial fission-related proteins. TBHQ activated the Parkin, Beclin, LC3II/I, and COXIV expression, while inhibiting the p62 expression. CSA reversed the effects of TBHQ on NSC proliferation and differentiation. After NSC(TBHQ) transplantation, it not only further extended the dwell time in the target quadrant and shorten the time and distance for finding the hidden platform, but also further decreased the Aβ and p-Tau/Tau levels, while increasing the expression of NeuN. The effects of NSC(TBHQ) transplantation on mitochondrial function were consistent with the in vitro experiments. CONCLUSIONS The study shows that NSC(TBHQ) intensifies the beneficial impact of NSCs transplantation on cognitive impairment and neuronal damage in AD models, likely due to TBHQ's role in promoting NSCs growth and differentiation via mitophagy, thus laying a theoretical foundation for improving NSCs transplantation for AD.
Collapse
Affiliation(s)
- Long Lin
- Department of Neurosurgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou 470000, Hainan, China
| | - Jiameng Li
- Department of Neurosurgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou 470000, Hainan, China
| | - Zhengtao Yu
- Department of Neurosurgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou 470000, Hainan, China
| | - Jun He
- Department of Neurosurgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou 470000, Hainan, China
| | - You Li
- Department of Neurosurgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou 470000, Hainan, China
| | - Junwen Jiang
- Department of Neurosurgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou 470000, Hainan, China
| | - Ying Xia
- Department of Neurosurgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou 470000, Hainan, China.
| |
Collapse
|
28
|
Cao ZL, Zhu LX, Wang HM, Zhu LJ. Microglial Regulation of Neural Networks in Neuropsychiatric Disorders. Neuroscientist 2025:10738584251316558. [PMID: 39932233 DOI: 10.1177/10738584251316558] [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: 02/23/2025]
Abstract
Microglia serve as vital innate immune cells in the central nervous system, playing crucial roles in the generation and development of brain neurons, as well as mediating a series of immune and inflammatory responses. The morphologic transitions of microglia are closely linked to their function. With the advent of single-cell sequencing technology, the diversity of microglial subtypes is increasingly recognized. The intricate interactions between microglia and neuronal networks have significant implications for psychiatric disorders and neurodegenerative diseases. A deeper investigation of microglia in neurologic diseases such as Alzheimer disease, depression, and epilepsy can provide valuable insights in understanding the pathogenesis of diseases and exploring novel therapeutic strategies, thereby addressing issues related to central nervous system disorders.
Collapse
Affiliation(s)
- Zi-Lin Cao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Xia Zhu
- Patent Examination Cooperation (JIANGSU) Center of the Patent Office, China National Intellectual Property Administration (CNIPA), Suzhou, China
| | - Hong-Mei Wang
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
29
|
Wu L, Sun Y, Yin Y, Wu Z, Liu R, Liu Y, Zhu Y, Shao M, Zhou H, Lu C, Zhang H. Lancao decoction in the treatment of alzheimer's disease via activating PI3K/AKT signaling to promote ERK involving in enhancing neuronal activities in the hippocampus. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119017. [PMID: 39528121 DOI: 10.1016/j.jep.2024.119017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/21/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Previous study has demonstrated lancao decoction (LC), a traditional Chinese medicine (TCM) fomula and recorded in "Huangdineijing", has a therapeutic effect on cognitive impairment (early clinical manifestations of alzheimer's disease (AD), which suggests that LC may have potential therapeutic advantages for AD. Whether LC has the therapeutic effect on AD and its potential mechanisms were still further indicated. AIM OF THE STUDY In this study, we aimed to uncover the potential advantage and neuronal mechanisms of LC in the treatment of AD in APP/PS1 mice in the hippocampus. METHODS AND MATERIALS We chose APP/PS1 mice to combing with behavioral tests including morris water maze (MWM) or y-maze to determine the role of LC in the therapeutic actions of AD. Network pharmacology was used to screen potential targets and pathways involving in LC's treatments of AD. Western blot was used to detect the phosphorylated expressions of proteins in hippocampus in APP/PS1 mice in the hippocampus. Pharmacological interventions were used to elucidate the relationship between the role of LC in the treatment of AD and the pathway, as well as the upstream and downstream interactions with neuronal activities. RESULTS According to our previous LC effective dose (2.5 g/kg), the dose was also able to significantly reduce the latency to the platform, and significantly increase the number of crossing times and time spend in the target quadrant in APP/PS1 mice in MWM, which was consistent with donepezil (DON) after 14 days chronic treatments. Network pharmacology showed that PI3K/AKT and MAPK pathways were closely associated with LC's treatments of AD, and protein autophosphorylation played a role in this process. The phosphorylated expressions of PI3K and AKT were obviously reduced in APP/PS1 mice in the hippocampus, which were both reversed by LC or DON. The phosphorylated expressions of MAPK including P38, JNK and ERK were also significantly reduced in APP/PS1 mice hippocampus, but only the phosphorylated expression of ERK was reversed by LC or DON. Inhibiting the activities of PI3K/AKT pathway by LY294002 blocked LC's improvement of behavioral deficits in APP/PS1 mice, including reducing latency to platform and increasing the number of crossings time in MWM in APP/PS1 mice, which also blunted LC's up-regulated phosphorylated expressions of PI3K, AKT and ERK in the hippocampus. Moreover, suppressing the activities of ERK by PD98059 also blocked LC's improvement of AD-related behavioral deficits including decreasing latency to new arm and increasing time in new arm in y-maze test, which also inhibited LC's enhancement of synaptic proteins (PSD95 and synapsin1) in the hippocampus and the number of EGR1-positive cells in the hippocampal dentate gyrus (DG). CONCLUSIONS Take together, our study revealed that LC had the therapeutic effects on AD by activating the PI3K/AKT pathway to enhance ERK activity and further strengthened neuronal activities in the hippocampus.
Collapse
Affiliation(s)
- Lei Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing, 210029, China
| | - Yan Sun
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ying Yin
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China
| | - Zhangjie Wu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China
| | - Ruiyi Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China
| | - Yuxin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing, 210029, China
| | - Yaping Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing, 210029, China
| | - Mengqi Shao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing, 210029, China
| | - Hang Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Danyang Hospital of Traditional Chinese Medicine, Zhenjiang, 212399, China
| | - Chao Lu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing, 210029, China.
| | - Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632, China; The Guangdong-Hongkong-Macau Joint Laboratory of Traditional Chinese Medicine Regulation of Brain-Periphery Homeostasis and Comprehensive Health, Guangzhou, 510632, China; Zhuhai Institute of Jinan University, Zhuhai, 519070, China.
| |
Collapse
|
30
|
Han S, Hwang J, Park T, Pyun J, Lee J, Park JS, Bice PJ, Liu S, Yun S, Jeong J, Risacher SL, Saykin AJ, Byun MS, Yi D, Sung J, Lee DY, Kim S, Nho K, Park YH. Transcriptome analysis of early- and late-onset Alzheimer's disease in Korean cohorts. Alzheimers Dement 2025; 21:e14563. [PMID: 39935412 PMCID: PMC11815242 DOI: 10.1002/alz.14563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 02/13/2025]
Abstract
INTRODUCTION The molecular mechanisms underlying early-onset Alzheimer's disease (EOAD) and late-onset Alzheimer's disease (LOAD) remain incompletely understood, particularly in Asian populations. METHODS RNA-sequencing was carried out on blood samples from 248 participants in the Seoul National University Bundang Hospital cohort to perform differential gene expression (DGE) and weighted gene co-expression network analysis. Findings were replicated in an independent Korean cohort (N = 275). RESULTS DGE analysis identified 18 and 88 dysregulated genes in EOAD and LOAD, respectively. Network analysis identified a LOAD-associated module showing a significant enrichment in pathways related to mitophagy, 5' adenosine monophosphate-activated protein kinase signaling, and ubiquitin-mediated proteolysis. In the replication cohort, downregulation of SMOX and PLVAP in LOAD was replicated, and the LOAD-associated module was highly preserved. In addition, SMOX and PLVAP were associated with brain amyloid beta deposition. DISCUSSION Our findings suggest distinct molecular signatures for EOAD and LOAD in a Korean population, providing deeper understanding of their diagnostic potential and molecular mechanisms. HIGHLIGHTS Analysis identified 18 and 88 dysregulated genes in early-onset Alzheimer's disease (EOAD) and late-onset Alzheimer's disease (LOAD), respectively. Expression levels of SMOX and PLVAP were downregulated in LOAD. Expression levels of SMOX and PLVAP were associated with brain amyloid beta deposition. Pathways including mitophagy and 5' adenosine monophosphate-activated protein kinase signaling were enriched in a LOAD module. A LOAD module was highly preserved across two independent cohorts.
Collapse
Affiliation(s)
- Sang‐Won Han
- Department of NeurologySoonchunhyang University Seoul HospitalSeoulRepublic of Korea
- Department of NeurologyChuncheon Sacred Heart Hospital, Hallym University College of MedicineChuncheon‐siRepublic of Korea
| | - Jiyun Hwang
- Genome and Health Big Data Laboratory, Graduate School of Public HealthSeoul National UniversitySeoulRepublic of Korea
| | - Tamina Park
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jung‐Min Pyun
- Department of NeurologySoonchunhyang University Seoul HospitalSeoulRepublic of Korea
| | - Joo‐Yeon Lee
- Genome and Health Big Data Laboratory, Graduate School of Public HealthSeoul National UniversitySeoulRepublic of Korea
- Institute of Health and EnvironmentsSeoul National UniversitySeoulRepublic of Korea
| | - Jeong Su Park
- Department of Laboratory MedicineSeoul National University Bundang Hospital, Seoul National University College of MedicineSeongnam‐siRepublic of Korea
| | - Paula J. Bice
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Shiwei Liu
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sunmin Yun
- Precision Medicine CenterSeoul National University Bundang HospitalSeongnam‐siRepublic of Korea
| | - Jibin Jeong
- Precision Medicine CenterSeoul National University Bundang HospitalSeongnam‐siRepublic of Korea
| | - Shannon L. Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andrew J. Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Min Soo Byun
- Department of PsychiatrySeoul National University College of MedicineSeoulRepublic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research CenterSeoul National UniversitySeoulRepublic of Korea
| | - Joohon Sung
- Genome and Health Big Data Laboratory, Graduate School of Public HealthSeoul National UniversitySeoulRepublic of Korea
- Institute of Health and EnvironmentsSeoul National UniversitySeoulRepublic of Korea
| | - Dong Young Lee
- Department of PsychiatrySeoul National University College of MedicineSeoulRepublic of Korea
| | - SangYun Kim
- Department of NeurologySeoul National University Bundang Hospital and Seoul National University College of MedicineSeongnam‐siRepublic of Korea
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
| | - Young Ho Park
- Department of NeurologySeoul National University Bundang Hospital and Seoul National University College of MedicineSeongnam‐siRepublic of Korea
| |
Collapse
|
31
|
Li SY, Gong XY, Ndikuryayo F, Yang WC. The emerging role of oxygen redox in pathological progression of disorders. Ageing Res Rev 2025; 104:102660. [PMID: 39805473 DOI: 10.1016/j.arr.2025.102660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and Huntington disease, pose serious threats to human health, leading to substantial economic burdens on society and families. Despite extensive research, the underlying mechanisms driving these diseases remain incompletely understood, impeding effective diagnosis and treatment. In recent years, growing evidence has highlighted the crucial role of oxidative stress in the pathogenesis of various neurodegenerative diseases. However, there is still a lack of comprehensive reviews that systematically summarize the impact of mitochondrial oxidative stress on neurodegenerative diseases. This review aims to address this gap by summarizing the molecular mechanisms by which mitochondrial oxidative stress promotes the initiation and progression of neurodegenerative disorders. Furthermore, it discusses the potential of antioxidant-based therapeutic strategies for the treatment of these diseases. By shedding light on the role of mitochondrial oxidative stress in neurodegenerative diseases, this review not only serves as a valuable reference for further research on the disease mechanisms, but also offers novel perspectives for the treatment of these disorders.
Collapse
Affiliation(s)
- Shuang-Yu Li
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, PR China
| | - Xue-Yan Gong
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, PR China
| | - Ferdinand Ndikuryayo
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, PR China
| | - Wen-Chao Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, PR China.
| |
Collapse
|
32
|
Cheng Y, Zhao A, Li Y, Li C, Miao X, Yang W, Wang Y. Roles of SIRT3 in cardiovascular and neurodegenerative diseases. Ageing Res Rev 2025; 104:102654. [PMID: 39755174 DOI: 10.1016/j.arr.2024.102654] [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: 11/20/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
Sirtuin-3 (SIRT3) in mitochondria has nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase activity. As such, SIRT3 is crucial in cardiovascular and neurodegenerative diseases. Advanced proteomics and transcriptomics studies have revealed that SIRT3 expression becomes altered when the heart or brain is affected by external stimuli or disease, such as diabetic cardiomyopathy, atherosclerosis, myocardial infarction, Alzheimer's disease, Huntington's disease, and Parkinson's disease. More specifically, SIRT3 participates in the development of these disorders through its deacetylase activity and in combination with downstream signaling pathways. The paper reviews SIRT3's expression changes, roles, and mechanisms associated with the development of cardiovascular and neurodegenerative diseases. Additionally, strategies targeting SIRT3 to treat or regulate cardiovascular and neurodegenerative disease development are discussed.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China; Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Anqi Zhao
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Ying Li
- Department of Medical Clinic, Jilin Women and Children Health Hospital, Changchun, Jilin, China
| | - Cheng Li
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiao Miao
- The Second Hosptial of Jilin University, Changchun, Jilin, China.
| | - Wanshan Yang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China.
| | - Yonggang Wang
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
33
|
Manna PR, Yang S, Manna C, Waters H, Islam MA, Reddy AP, Rawat P, Reddy PH. Steroidogenic acute regulatory protein mediated variations of gender-specific sex neurosteroids in Alzheimer's disease: Relevance to hormonal and neuronal imbalance. Neurosci Biobehav Rev 2025; 169:105969. [PMID: 39631487 DOI: 10.1016/j.neubiorev.2024.105969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
The steroidogenic acute regulatory (StAR) protein mediates the rate-liming step in neuro/steroid biosynthesis. Multifaceted and delicate changes during aging, disrupting hormonal and neuronal homeostasis, constitute human senescence, an inevitable phenomenon that attributes to increased morbidity and mortality. Aging, along with progressive decreases in bioactive neurosteroids, is the primary risk factor for Alzheimer's disease (AD), which preferentially impacts two-thirds of women and one-third of men. AD is neuropathologically characterized by the accumulation of extracellular amyloid-β and intracellular phosphorylated Tau containing neurofibrillary tangles, resulting in dementia. Postmortem brains pertaining to gender-specific AD patients exhibit varied suppression of StAR and sex neurosteroid levels compared with age-matched cognitively healthy subjects, in which the attenuation of StAR is inversely correlated with the AD pathological markers. Interestingly, retinoid signaling upregulates StAR-motivated neurosteroid biosynthesis and reinstates various neurodegenerative vulnerabilities that promote AD pathogenesis. This review summarizes current understanding of StAR-driven alterations of sex neurosteroids in gender-specific AD risks and provides biochemical and molecular insights into therapeutic interventions for preventing and/or alleviating dementia for healthy aging.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Chayan Manna
- Baylor College of Medicine, Ben Taub Research Center, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Hope Waters
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Priyanka Rawat
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
34
|
Wen P, Sun Z, Gou F, Wang J, Fan Q, Zhao D, Yang L. Oxidative stress and mitochondrial impairment: Key drivers in neurodegenerative disorders. Ageing Res Rev 2025; 104:102667. [PMID: 39848408 DOI: 10.1016/j.arr.2025.102667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Mitochondrial dysfunction and oxidative stress are critical factors in the pathogenesis of neurodegenerative diseases. The complex interplay between these factors exacerbates neuronal damage and accelerates disease progression. In neurodegenerative diseases, mitochondrial dysfunction impairs ATP production and promotes the generation of reactive oxygen species (ROS). The accumulation of ROS further damages mitochondrial DNA, proteins, and lipids, creating a vicious cycle of oxidative stress and mitochondrial impairment. This review aims to elucidate the mechanisms by which mitochondrial dysfunction and oxidative stress lead to neurodegeneration, and to highlight potential therapeutic targets to mitigate their harmful effects.
Collapse
Affiliation(s)
- Pei Wen
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhixin Sun
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Fengting Gou
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jingjing Wang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qing Fan
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Deming Zhao
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lifeng Yang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
35
|
Zhao W, Yang R, Meng X, Xu SQ, Li MM, Hao ZC, Wang SY, Jiang YK, Naseem A, Chen QS, Zhang LL, Kuang HX, Yang BY, Liu Y. Panax quinquefolium saponins protects neuronal activity by promoting mitophagy in both in vitro and in vivo models of Alzheimer's disease. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119250. [PMID: 39681202 DOI: 10.1016/j.jep.2024.119250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In the realm of traditional Chinese medicine, Panax quinquefolius L. has garnered significant attention for its potential to treat various ailments associated with deficiencies, including qi, blood, and kidneys. As its primary bioactive constituent, Panax quinquefolius saponins (PQS) have the potential therapeutic role of Alzheimer's disease (AD) treatment, but with unclear mechanisms of action. Meanwhile, AD is considered as a common dementia disease with kidney insufficiency and deficiency by traditional medicine, and often accompanied by autophagy in modern medical research. AIM OF THE STUDY This study aimed to investigate the therapeutic effects of PQS on AD through the regulation of mitophagy. MATERIALS AND METHODS The chemical constituents of PQS were characterized using the UPLC-QTOF-MS technique. After that, the HT22 cell line was used to establish the D-galactose-induced cell model, and the SAMP8 mice model of AD was also employed. Cell viability was assessed using the CCK-8 assay, ROS detection, JC-1 staining, Mito-tracker Red and LC3 staining, and Mito-tracker Green and Lyso-tracker Red staining were used to assess levels of mitophagy. The Morris Water Maze (MWM) was used for the experimental evaluation of learning and memory abilities in mice. Subsequently, the mechanism was studied by pathological staining and western blotting. RESULTS Fifty-eight triterpenoid saponins were identified from PQS, and PQS alleviated D-galactose-induced HT22 cell death and increased intracellular levels of mitochondrial autophagy-related factors. In vivo, PQS significantly improved cognitive deficits and mitigated AD-like pathological features by activating the mitophagy mechanism. Furthermore, PQS may promote Pink1/Parkin-mediated mitophagy by activating the AMPK/mTOR/ULK1/DRP1 and SIRT1/PGC-1α pathways. CONCLUSION In conclusion, PQS have demonstrated the potential to mitigate mitochondrial dysfunction and enhance cognitive function in AD through the activation of mitophagy. This promising strategy holds great promise for the treatment of AD.
Collapse
Affiliation(s)
- Wei Zhao
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Rui Yang
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Xin Meng
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Shi-Qi Xu
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Meng-Meng Li
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Zhi-Chao Hao
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Si-Yi Wang
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Yi-Kai Jiang
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Anam Naseem
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Qing-Shan Chen
- Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), Heilongjiang, Harbin, 150040, People's Republic of China.
| | - Li-Li Zhang
- Traditional Chinese Medicine (TCM) Biological Genetics (Heilongjiang Province Double First-class Construction Interdiscipline), Heilongjiang, Harbin, 150040, People's Republic of China.
| | - Hai-Xue Kuang
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Bing-You Yang
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| | - Yan Liu
- Heilongjiang University of Chinese Medicine, Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education Heilongjiang Touyan Innovation Team Program, Harbin, 150040, People's Republic of China.
| |
Collapse
|
36
|
Zhang X, Li J, Fu M, Geng X, Hu J, Tang KJ, Chen P, Zou J, Liu X, Zeng B. Dysfunction in mitochondrial electron transport chain drives the pathogenesis of pulmonary arterial hypertension: insights from a multi-omics investigation. Respir Res 2025; 26:29. [PMID: 39833797 PMCID: PMC11749457 DOI: 10.1186/s12931-025-03099-8] [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: 11/28/2023] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a progressive disorder that can lead to right ventricular failure and severe consequences. Despite extensive efforts, limited progress has been made in preventing the progression of PAH. Mitochondrial dysfunction is implicated in the development of PAH, but the key mitochondrial functional alterations in the pathogenesis have yet to be elucidated. METHODS We integrated three microarray datasets from the Gene Expression Omnibus (GEO), including 222 lung samples (164 PAH, 58 controls), for differential expression and functional enrichment analyses. Machine learning identified key mitochondria-related signaling pathways. PAH and control lung tissue samples were collected, and transcriptomic and metabolomic profiling were performed. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis investigated shared pathways, and canonical correlation analysis assessed gene-metabolite relationships. RESULTS In the GEO datasets, mitochondria-related signaling pathways were significantly enriched in PAH samples, in particular the electron transport chain (ETC) in mitochondrial oxidative phosphorylation system. Notably, the electron transport from cytochrome c to oxygen in ETC was identified as the most crucial mitochondria-related pathway, which was down-regulated in PAH samples. Transcriptomic profiling of the clinical lung tissue analysis identified 14 differentially expressed genes (DEGs) related to mitochondrial function. Metabolomic analysis revealed three differential metabolites in PAH samples: increased 3-phenyllactic acid and ADP, and decreased citric acid. Mitochondria-related genes highly correlated with these metabolites included KIT, OTC, CAMK2A, and CHRNA1. CONCLUSIONS Down-regulation of electron transport from cytochrome c to oxygen in mitochondrial ETC and disruption of the citric acid cycle homeostasis may contribute to PAH pathogenesis. 3-phenyllactic acid emerges as a potential novel diagnostic biomarker for PAH. These findings offer insights for developing novel PAH therapies and diagnostics.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jieling Li
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Minyi Fu
- Surgical and Anesthesia Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xijie Geng
- Surgical and Anesthesia Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Junjie Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ke-Jing Tang
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pan Chen
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianyong Zou
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Xiaoman Liu
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Bo Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
37
|
Wang S, Liao Z, Zhang Q, Han X, Liu C, Wang J. Mitochondrial dysfunction in Alzheimer's disease: a key frontier for future targeted therapies. Front Immunol 2025; 15:1484373. [PMID: 39877373 PMCID: PMC11772192 DOI: 10.3389/fimmu.2024.1484373] [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: 08/21/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, accounting for approximately 70% of dementia cases worldwide. Patients gradually exhibit cognitive decline, such as memory loss, aphasia, and changes in personality and behavior. Research has shown that mitochondrial dysfunction plays a critical role in the onset and progression of AD. Mitochondrial dysfunction primarily leads to increased oxidative stress, imbalances in mitochondrial dynamics, impaired mitophagy, and mitochondrial genome abnormalities. These mitochondrial abnormalities are closely associated with amyloid-beta and tau protein pathology, collectively accelerating the neurodegenerative process. This review summarizes the role of mitochondria in the development of AD, the latest research progress, and explores the potential of mitochondria-targeted therapeutic strategies for AD. Targeting mitochondria-related pathways may significantly improve the quality of life for AD patients in the future.
Collapse
Affiliation(s)
- Shuguang Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zuning Liao
- Department of Neurology, Fourth People’s Hospital of Jinan, Jinan, China
| | - Qiying Zhang
- Department of Internal Medicine, Jinan Municipal Government Hospital, Jinan, China
| | - Xinyuan Han
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changqing Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jin Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
38
|
Yu R, Zhang H, Chen R, Lin Y, Xu J, Fang Z, Ru Y, Fan C, Wu G. Fecal Microbiota Transplantation from Methionine-Restricted Diet Mouse Donors Improves Alzheimer's Learning and Memory Abilities Through Short-Chain Fatty Acids. Foods 2025; 14:101. [PMID: 39796390 PMCID: PMC11720665 DOI: 10.3390/foods14010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Alzheimer's disease (AD) is marked by impaired cognitive functions, particularly in learning and memory, owing to complex and diverse mechanisms. Methionine restriction (MR) has been found to exert a mitigating effect on brain oxidative stress to improve AD. However, the bidirectional crosstalk between the gut and brain through which MR enhances learning and memory in AD, as well as the effects of fecal microbiota transplantation (FMT) from MR mice on AD mice, remains underexplored. In this study, APP/PS1 double transgenic AD mice were used and an FMT experiment was conducted. 16S rRNA gene sequencing, targeted metabolomics, and microbial metabolite short-chain fatty acids (SCFAs) of feces samples were analyzed. The results showed that MR reversed the reduction in SCFAs induced by AD, and further activated the free fatty acid receptors, FFAR2 and FFAR3, as well as the transport protein MCT1, thereby signaling to the brain to mitigate inflammation and enhance the learning and memory capabilities. Furthermore, the FMT experiment from methionine-restricted diet mouse donors showed that mice receiving FMT ameliorated Alzheimer's learning and memory ability through SCFAs. This study offers novel non-pharmaceutical intervention strategies for AD prevention.
Collapse
Affiliation(s)
- Run Yu
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Haimeng Zhang
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Rui Chen
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yangzhuo Lin
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Jingxuan Xu
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Ziyang Fang
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yuehang Ru
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Chenhan Fan
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Guoqing Wu
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| |
Collapse
|
39
|
Olesen MA, Villavicencio-Tejo F, Cuevas-Espinoza V, Quintanilla RA. Unknown roles of tau pathology in neurological disorders. Challenges and new perspectives. Ageing Res Rev 2025; 103:102594. [PMID: 39577774 DOI: 10.1016/j.arr.2024.102594] [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: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Aging presents progressive changes that increase the susceptibility of the central nervous system (CNS) to suffer neurological disorders (NDs). Several studies have reported that an aged brain suffering from NDs shows the presence of pathological forms of tau protein, a microtubule accessory protein (MAP) critical for neuronal function. In this context, accumulative evidence has shown a pivotal contribution of pathological forms of tau to Alzheimer's disease (AD) and tauopathies. However, current investigations have implicated tau toxicity in other NDs that affect the central nervous system (CNS), including Parkinson's disease (PD), Huntington's disease (HD), Traumatic brain injury (TBI), Multiple sclerosis (MS), and Amyotrophic lateral sclerosis (ALS). These diseases are long-term acquired, affecting essential functions such as motor movement, cognition, hearing, and vision. Previous evidence indicated that toxic forms of tau do not have a critical contribution to the genesis or progression of these diseases. However, recent studies have shown that these tau forms contribute to neuronal dysfunction, inflammation, oxidative damage, and mitochondrial impairment events that contribute to the pathogenesis of these NDs. Recent studies have suggested that these neuropathologies could be associated with a prion-like behavior of tau, which induces a pathological dissemination of these toxic protein forms to different brain areas. Moreover, it has been suggested that this toxic propagation of tau from neurons into neighboring cells impairs the function of glial cells, oligodendrocytes, and endothelial cells by affecting metabolic function and mitochondrial health and inducing oxidative damage by tau pathology. Therefore, in this review, we will discuss current evidence demonstrating the critical role of toxic tau forms on NDs not related to AD and how its propagation and induced-bioenergetics failure may contribute to the pathogenic mechanism present in these NDs.
Collapse
Affiliation(s)
- Margrethe A Olesen
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Francisca Villavicencio-Tejo
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Víctor Cuevas-Espinoza
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile.
| |
Collapse
|
40
|
Siwach A, Patel H, Khairnar A, Parekh P. Molecular Symphony of Mitophagy: Ubiquitin-Specific Protease-30 as a Maestro for Precision Management of Neurodegenerative Diseases. CNS Neurosci Ther 2025; 31:e70192. [PMID: 39840724 PMCID: PMC11751875 DOI: 10.1111/cns.70192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/24/2024] [Accepted: 12/07/2024] [Indexed: 01/30/2025] Open
Abstract
INTRODUCTION Mitochondrial dysfunction stands as a pivotal feature in neurodegenerative disorders, spurring the quest for targeted therapeutic interventions. This review examines Ubiquitin-Specific Protease 30 (USP30) as a master regulator of mitophagy with therapeutic promise in Alzheimer's disease (AD) and Parkinson's disease (PD). USP30's orchestration of mitophagy pathways, encompassing PINK1-dependent and PINK1-independent mechanisms, forms the crux of this exploration. METHOD A systematic literature search was conducted in PubMed, Scopus, and Web of Science, selecting studies that investigated USP's function, inhibitor design, or therapeutic efficacy in AD and PD. Inclusion criteria encompassed mechanistic and preclinical/clinical data, while irrelevant or duplicate references were excluded. Extracted findings were synthesized narratively. RESULTS USP30 modulates interactions with translocase of outer mitochondrial membrane (TOM) 20, mitochondrial E3 ubiquitin protein ligase 1 (MUL1), and Parkin, thus harmonizing mitochondrial quality control. Emerging novel USP30 inhibitors, racemic phenylalanine derivatives, N-cyano pyrrolidine, and notably, benzosulphonamide class compounds, restore mitophagy, and reduce neurodegenerative phenotypes across diverse models with minimal off-target effects. Modulation of other USPs also influences neurodegenerative disease pathways, offering additional therapeutic avenues. CONCLUSIONS In highlighting the nuanced regulation of mitophagy by USP30, this work heralds a shift toward more precise and effective treatments, paving the way for a new era in the clinical management of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ankit Siwach
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)AhmedabadGujaratIndia
- School of Pharmaceutical SciencesJaipur National UniversityJaipurRajasthanIndia
| | - Harit Patel
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)AhmedabadGujaratIndia
| | - Amit Khairnar
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)AhmedabadGujaratIndia
- Department of Physiology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
- International Clinical Research Center (ICRC)St. Anne's University HospitalBrnoCzech Republic
- International Clinical Research Center (ICRC), Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Pathik Parekh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on AgingNational Institutes of HealthBaltimoreUSA
| |
Collapse
|
41
|
Bondy SC, Wu M. The Critical Role of Autophagy and Phagocytosis in the Aging Brain. Int J Mol Sci 2024; 26:57. [PMID: 39795916 PMCID: PMC11720579 DOI: 10.3390/ijms26010057] [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: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
As the organism ages, there is a decline in effective energy supply, and this retards the ability to elaborate new proteins. The consequences of this are especially marked in the gradual decline in brain function. The senescence of cells and their constituent organelles is ultimately the cause of aging of the entire nervous system. What is less immediately obvious is that brain aging is also accompanied by the failure of catabolic events that lead to the removal of non-functional cells and ineffective subcellular components. The removal of non-working cellular and subcellular elements within the brain is essential in order to allow the appearance of fresh cells and organelles with a full range of capacities. Thus, the maintenance of operative mechanisms for the dispersal of failed tissue components is important, and its diminished capacity with aging is a significant contributory factor to the onset and progression of age-related neurological disorder. This report discusses the mechanisms underlying autophagy and phagocytosis and how these can be adversely modulated as aging proceeds. The means by which the effective recycling of cellular components may be reinstated in the aged brain are considered.
Collapse
Affiliation(s)
- Stephen C. Bondy
- Department of Occupational and Environmental Health and Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Meixia Wu
- Evergreen World ADHC, Westminster, CA 92844, USA;
| |
Collapse
|
42
|
Di Credico A, Gaggi G, Bibbò S, Pilato S, Moffa S, Di Giacomo S, Siani G, Fontana A, Konstantinidou F, Donato M, Stuppia L, Gatta V, Di Baldassarre A, Ghinassi B. Exploring Potential Impact of Graphene Oxide and Graphene Oxide-Polyethylenimine on Biological Behavior of Human Amniotic Fluid-Derived Stem Cells. Int J Mol Sci 2024; 25:13598. [PMID: 39769359 PMCID: PMC11678234 DOI: 10.3390/ijms252413598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Regenerative medicine and tissue engineering aim to restore or replace impaired organs and tissues using cell transplantation supported by scaffolds. Recently scientists are focusing on developing new biomaterials that optimize cellular attachment, migration, proliferation, and differentiation. Nanoparticles, such as graphene oxide (GO), have emerged as versatile materials due to their high surface-to-volume ratio and unique chemical properties, such as electrical conductivity and flexibility. However, GO faces challenges such as cytotoxicity at high concentrations, a negative surface charge, and potential inflammatory responses; for these reasons, variations in synthesis have been studied. A GO derivative, Graphene Oxide-Polyethylenimine (GO-PEI), shows controlled porosity and structural definition, potentially offering better support for cell growth. Human amniotic fluid stem cells (hAFSCs) are a promising candidate for regenerative medicine due to their ability to differentiate into mesodermic and ectodermic lineages, their non-immunogenic nature, and ease of isolation. This study investigates the effects of GO and GO-PEI on hAFSCs, focusing on the effects on adhesion, proliferation, and metabolic features. Results indicate that GO-PEI restores cell proliferation and mitochondrial activity to control levels, with respect to GO that appeared less biocompatible. Both materials also influence the miRNA cargo of hAFSC-derived microvesicles, potentially influencing also cell-to-cell communication.
Collapse
Affiliation(s)
- Andrea Di Credico
- Department of Medicine and Aging Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (G.G.); (S.B.)
- Cell Reprogramming and Differentiation Lab, “G. d’Annunzio University” of Chieti-Pescara, 66100 Chieti, Italy;
- UdA-Tech Lab, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (A.F.)
| | - Giulia Gaggi
- Department of Medicine and Aging Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (G.G.); (S.B.)
- Cell Reprogramming and Differentiation Lab, “G. d’Annunzio University” of Chieti-Pescara, 66100 Chieti, Italy;
- UdA-Tech Lab, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (A.F.)
| | - Sandra Bibbò
- Department of Medicine and Aging Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (G.G.); (S.B.)
- Cell Reprogramming and Differentiation Lab, “G. d’Annunzio University” of Chieti-Pescara, 66100 Chieti, Italy;
| | - Serena Pilato
- UdA-Tech Lab, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (A.F.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.M.); (S.D.G.); (G.S.)
| | - Samanta Moffa
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.M.); (S.D.G.); (G.S.)
| | - Stefano Di Giacomo
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.M.); (S.D.G.); (G.S.)
| | - Gabriella Siani
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.M.); (S.D.G.); (G.S.)
| | - Antonella Fontana
- UdA-Tech Lab, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (A.F.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.M.); (S.D.G.); (G.S.)
| | - Fani Konstantinidou
- Department of Neuroscience, Imaging and Clinical Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.K.); (M.D.); (L.S.); (V.G.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Marisa Donato
- Department of Neuroscience, Imaging and Clinical Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.K.); (M.D.); (L.S.); (V.G.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Neuroscience, Imaging and Clinical Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.K.); (M.D.); (L.S.); (V.G.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Neuroscience, Imaging and Clinical Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (F.K.); (M.D.); (L.S.); (V.G.)
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (G.G.); (S.B.)
- Cell Reprogramming and Differentiation Lab, “G. d’Annunzio University” of Chieti-Pescara, 66100 Chieti, Italy;
- UdA-Tech Lab, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (A.F.)
| | - Barbara Ghinassi
- Cell Reprogramming and Differentiation Lab, “G. d’Annunzio University” of Chieti-Pescara, 66100 Chieti, Italy;
- UdA-Tech Lab, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.P.); (A.F.)
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
43
|
Ren N, Zhang H, Li T, Ji H, Zhang Z, Wu H. ATP5J regulates microglial activation via mitochondrial dysfunction, exacerbating neuroinflammation in intracerebral hemorrhage. Front Immunol 2024; 15:1509370. [PMID: 39735538 PMCID: PMC11671693 DOI: 10.3389/fimmu.2024.1509370] [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: 10/10/2024] [Accepted: 12/02/2024] [Indexed: 12/31/2024] Open
Abstract
Microglial-mediated neuroinflammation is crucial in the pathophysiological mechanisms of secondary brain injury (SBI) following intracerebral hemorrhage (ICH). Mitochondria are central regulators of inflammation, influencing key pathways such as alternative splicing, and play a critical role in cell differentiation and function. Mitochondrial ATP synthase coupling factor 6 (ATP5J) participates in various pathological processes, such as cell proliferation, migration, and inflammation. However, the role of ATP5J in microglial activation and neuroinflammation post-ICH is poorly understood. This study aimed to investigate the effects of ATP5J on microglial activation and subsequent neuroinflammation in ICH and to elucidate the underlying mechanisms. We observed that ATP5J was upregulated in microglia after ICH. AAV9-mediated ATP5J overexpression worsened neurobehavioral deficits, disrupted the blood-brain barrier, and increased brain water content in ICH mice. Conversely, ATP5J knockdown ameliorated these effects. ATP5J overexpression also intensified microglial activation, neuronal apoptosis, and inflammatory responses in surrounding tissues post-ICH. ATP5J impaired microglial dynamics and reduced the proliferation and migration of microglia to injury sites. We used oxyhemoglobin (OxyHb) to stimulate BV2 cells and model ICH in vitro. Further in vitro studies showed that ATP5J overexpression enhanced OxyHb-induced microglial functional transformation. Mechanistically, ATP5J silencing reversed dynamin-related protein 1 (Drp1) and mitochondrial fission 1 protein (Fis1) upregulation in microglia post-OxyHb induction; reduced mitochondrial overdivision, excessive mitochondrial permeability transition pore opening, and reactive oxygen species production; restored normal mitochondrial ridge morphology; and partially restored mitochondrial respiratory electron transport chain activity. ATP5J silencing further alleviated OxyHb-induced mitochondrial dysfunction by regulating mitochondrial metabolism. Our results indicate that ATP5J is a key factor in regulating microglial functional transformation post-ICH by modulating mitochondrial dysfunction and metabolism, thereby positively regulate neuroinflammation. By inhibiting ATP5J, SBI following ICH could be prevented. Therefore, ATP5J could be a candidate for molecular and therapeutic target exploration to alleviate neuroinflammation post-ICH.
Collapse
Affiliation(s)
| | | | | | | | - Zhen Zhang
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin, China
| | - He Wu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
44
|
Lv D, Feng P, Guan X, Liu Z, Li D, Xue C, Bai B, Hölscher C. Neuroprotective effects of GLP-1 class drugs in Parkinson's disease. Front Neurol 2024; 15:1462240. [PMID: 39719978 PMCID: PMC11667896 DOI: 10.3389/fneur.2024.1462240] [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: 07/09/2024] [Accepted: 11/14/2024] [Indexed: 12/26/2024] Open
Abstract
Parkinson's disease (PD) is a chronic, progressive neurological disorder primarily affecting motor control, clinically characterized by resting tremor, bradykinesia, rigidity, and other symptoms that significantly diminish the quality of life. Currently, available treatments only alleviate symptoms without halting or delaying disease progression. There is a significant association between PD and type 2 diabetes mellitus (T2DM), possibly due to shared pathological mechanisms such as insulin resistance, chronic inflammation, and mitochondrial dysfunction. PD is caused by a deficiency of dopamine, a neurotransmitter in the brain that plays a critical role in the control of movement. Glucose metabolism and energy metabolism disorders also play an important role in the pathogenesis of PD. This review investigates the neuroprotective mechanisms of glucagon-like peptide-1 (GLP-1) and its receptor agonists, offering novel insights into potential therapeutic strategies for PD. GLP-1 class drugs, primarily used in diabetes management, show promise in addressing PD's underlying pathophysiological mechanisms, including energy metabolism and neuroprotection. These drugs can cross the blood-brain barrier, improve insulin resistance, stabilize mitochondrial function, and enhance neuronal survival and function. Additionally, they exhibit significant anti-inflammatory and antioxidative stress effects, which are crucial in neurodegenerative diseases like PD. Research indicates that GLP-1 receptor agonists could improve both motor and cognitive symptoms in PD patients, marking a potential breakthrough in PD treatment and prevention. Further exploration of GLP-1's molecular mechanisms in PD could provide new preventive and therapeutic approaches, especially for PD patients with concurrent T2DM. By targeting both metabolic and neurodegenerative pathways, GLP-1 receptor agonists represent a multifaceted approach to PD treatment, offering hope for better disease management and improved patient outcomes.
Collapse
Affiliation(s)
- Dongliang Lv
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Peng Feng
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Xueying Guan
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Zhaona Liu
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Dongfang Li
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Cunshui Xue
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Bo Bai
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Christian Hölscher
- Henan Academy of Innovations in Medical Science, Brain Institute, Zhengzhou, China
| |
Collapse
|
45
|
Yuan Y, Zhao G, Zhao Y. Dysregulation of energy metabolism in Alzheimer's disease. J Neurol 2024; 272:2. [PMID: 39621206 PMCID: PMC11611936 DOI: 10.1007/s00415-024-12800-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 12/06/2024]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. Its etiology and associated mechanisms are still unclear, which largely hinders the development of AD treatment strategies. Many studies have shown that dysregulation of energy metabolism in the brain of AD is closely related to disease development. Dysregulation of brain energy metabolism in AD brain is associated with reduced glucose uptake and utilization, altered insulin signaling pathways, and mitochondrial dysfunction. In this study, we summarized the relevant pathways and mechanisms regarding the dysregulation of energy metabolism in AD. In addition, we highlight the possible role of mitochondrial dysfunction as a central role in the AD process. A deeper understanding of the relationship between energy metabolism dysregulation and AD may provide new insights for understanding learning memory impairment in AD patients and in improving AD prevention and treatment.
Collapse
Affiliation(s)
- Yue Yuan
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, 130061, China
| | - Gang Zhao
- China Resources Pharmaceutical Commercial Group, Beijing, China
| | - Yang Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, 130061, China.
| |
Collapse
|
46
|
Tian Z, Zhang Q, Wang L, Li M, Li T, Wang Y, Cao Z, Jiang X, Luo P. Progress in the mechanisms of pain associated with neurodegenerative diseases. Ageing Res Rev 2024; 102:102579. [PMID: 39542176 DOI: 10.1016/j.arr.2024.102579] [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: 09/04/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Neurodegenerative diseases (NDDs) represent a class of neurological disorders characterized by the progressive degeneration or loss of neurons, impacting millions of individuals globally. In addition to the typical manifestations, pain is a prevalent symptom associated with NDDs, seriously impacting the quality of life for patients. The pathogenesis of pain associated with NDDs is intricate and multifaceted. Currently, the clinical management of NDDs-related pain symptoms predominantly relies on conventional pharmacological agents or physical therapy. However, these approaches often fail to produce satisfactory outcomes. This article summarizes the underlying mechanisms of major NDDs-associated pain: Neuroinflammation, Brain and spinal cord dysfunctions, Mitochondrial dysfunction, Risk gene and pathological protein, as well as Receptor, channel, and neurotransmitter. While numerous studies have investigated the downstream pathological processes associated with these mechanisms, there remains a significant gap in identifying the key initiating factors. Specifically, there is insufficient evidence for the upstream elements that activate microglia and astrocytes in neuroinflammation leading to pain in NDDs. Likewise, there is an absence of upstream factors elucidating how dysfunctions in the brain and spinal cord, as well as mitochondrial impairments, contribute to the development of pain. Furthermore, the specific mechanisms through which hallmark pathological proteins related to NDDs contribute to these pathological processes remain inadequately understood. The objective of this article is to synthesize the existing mechanisms underlying pain associated with NDDs, including Alzheimer's disease, Parkinson's disease, Huntington's disease, Schizophrenia, Amyotrophic lateral sclerosis, and Multiple sclerosis, while also identifying gaps and deficiencies in these mechanisms. This paper offers insights for future research trajectories. Given the intricate pathogenesis of NDDs-related pain, it emphasizes that a promising short-term strategy is combination therapy-intervening concurrently in multiple pathological processes-akin to the cocktail approach utilized in treating acquired immunodeficiency syndrome (AIDS). For long-term advancements, achieving breakthroughs in the treatment of the NDDs themselves will remain essential for alleviating accompanying pain symptoms.
Collapse
Affiliation(s)
- Zhicheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Qi Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; The Fifth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ling Wang
- Xi'an Children's Hospital, Xi'an 710002, China
| | - Mengxiang Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; The Fifth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Tianjing Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yujie Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Zixuan Cao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; The Sixth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
47
|
Maggiore A, Latina V, D'Erme M, Amadoro G, Coccurello R. Non-canonical pathways associated to Amyloid beta and tau protein dyshomeostasis in Alzheimer's disease: A narrative review. Ageing Res Rev 2024; 102:102578. [PMID: 39542177 DOI: 10.1016/j.arr.2024.102578] [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: 09/18/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Alzheimer's Disease (AD) is the most common form of dementia among elderly people. This disease imposes a significant burden on the healthcare system, society, and economy due to the increasing global aging population. Current trials with drugs or bioactive compounds aimed at reducing cerebral Amyloid beta (Aβ) plaques and tau protein neurofibrillary tangles, which are the two main hallmarks of this devastating neurodegenerative disease, have not provided significant results in terms of their neuropathological outcomes nor met the expected clinical end-points. Ageing, genetic and environmental risk factors, along with different clinical symptoms suggest that AD is a complex and heterogeneous disorder with multiple interconnected pathological pathways rather than a single disease entity. In the present review, we highlight and discuss various non-canonical, Aβ-independent mechanisms, like gliosis, unhealthy dietary intake, lipid and sugar signaling, and cerebrovascular damage that contribute to the onset and development of AD. We emphasize that challenging the traditional "amyloid cascade hypothesis" may improve our understanding of this age-related complex syndrome and help fight the progressive cognitive decline in AD.
Collapse
Affiliation(s)
- Anna Maggiore
- Department of Biochemical Sciences, Sapienza University, P.le Aldo Moro 5, Rome 00185, Italy; Department of Brain Sciences, Imperial College, London, UK
| | - Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, Rome 00161, Italy; Institute of Translational Pharmacology (IFT) CNR, Via Fosso del Cavaliere 100, Rome 00133, Italy
| | - Maria D'Erme
- Department of Biochemical Sciences, Sapienza University, P.le Aldo Moro 5, Rome 00185, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, Rome 00161, Italy; Institute of Translational Pharmacology (IFT) CNR, Via Fosso del Cavaliere 100, Rome 00133, Italy.
| | - Roberto Coccurello
- Institute for Complex System (ISC) CNR, Via dei Taurini 19, Rome 00185, Italy; IRCSS Santa Lucia Foundation, European Center for Brain Research, Via Fosso del Fiorano 64-65, Rome 00143, Italy.
| |
Collapse
|
48
|
Zhou YT, Li S, Du SL, Zhao JH, Cai YQ, Zhang ZQ. The multifaceted role of macrophage mitophagy in SiO 2-induced pulmonary fibrosis: A brief review. J Appl Toxicol 2024; 44:1854-1867. [PMID: 38644760 DOI: 10.1002/jat.4612] [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: 02/16/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/23/2024]
Abstract
Prolonged exposure to environments with high concentrations of crystalline silica (CS) can lead to silicosis. Macrophages play a crucial role in the pathogenesis of silicosis. In the process of silicosis, silica (SiO2) invades alveolar macrophages (AMs) and induces mitophagy which usually exists in three states: normal, excessive, and/or deficiency. Different mitophagy states lead to corresponding toxic responses, including successful macrophage repair, injury, necrosis, apoptosis, and even pulmonary fibrosis. This is a complex process accompanied by various cytokines. Unfortunately, the details have not been fully systematically summarized. Therefore, it is necessary to elucidate the role of macrophage mitophagy in SiO2-induced pulmonary fibrosis by systematic analysis on the literature reports. In this review, we first summarized the current data on the macrophage mitophagy in the development of SiO2-induced pulmonary fibrosis. Then, we introduce the molecular mechanism on how SiO2-induced mitophagy causes pulmonary fibrosis. Finally, we focus on introducing new therapies based on newly developed mitophagy-inducing strategies. We conclude that macrophage mitophagy plays a multifaceted role in the progression of SiO2-induced pulmonary fibrosis, and reprogramming the macrophage mitophagy state accordingly may be a potential means of preventing and treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Yu-Ting Zhou
- Department of Public Health, Shandong First Medical University, Jinan, China
- Department of Public Health, Jining Medical University, Jining, China
| | - Shuang Li
- Department of Public Health, Jining Medical University, Jining, China
| | - Shu-Ling Du
- Department of Public Health, Jining Medical University, Jining, China
| | - Jia-Hui Zhao
- Department of Public Health, Jining Medical University, Jining, China
| | | | - Zhao-Qiang Zhang
- Department of Public Health, Jining Medical University, Jining, China
| |
Collapse
|
49
|
Behl T, Kyada A, Roopashree R, Nathiya D, Arya R, Kumar MR, Khalid M, Gulati M, Sachdeva M, Fareed M, Patra PK, Agrawal A, Wal P, Gasmi A. Epigenetic biomarkers in Alzheimer's disease: Diagnostic and prognostic relevance. Ageing Res Rev 2024; 102:102556. [PMID: 39490904 DOI: 10.1016/j.arr.2024.102556] [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: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) is a leading cause of cognitive decline in the aging population, presenting a critical need for early diagnosis and effective prognostic tools. Epigenetic modifications, including DNA methylation, histone modifications, and non-coding RNAs, have emerged as promising biomarkers for AD due to their roles in regulating gene expression and potential for reversibility. This review examines the current landscape of epigenetic biomarkers in AD, emphasizing their diagnostic and prognostic relevance. DNA methylation patterns in genes such as APP, PSEN1, and PSEN2 are highlighted for their strong associations with AD pathology. Alterations in DNA methylation at specific CpG sites have been consistently observed in AD patients, suggesting their utility in early detection. Histone modifications, such as acetylation and methylation, also play a crucial role in chromatin remodelling and gene expression regulation in AD. Dysregulated histone acetylation and methylation have been linked to AD progression, making these modifications valuable biomarkers. Non-coding RNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), further contribute to the epigenetic regulation in AD. miRNAs can modulate gene expression post-transcriptionally and have been found in altered levels in AD, while lncRNAs can influence chromatin structure and gene expression. The presence of these non-coding RNAs in biofluids like blood and cerebrospinal fluid positions them as accessible and minimally invasive biomarkers. Technological advancements in detecting and quantifying epigenetic modifications have propelled the field forward. Techniques such as next-generation sequencing, bisulfite sequencing, and chromatin immunoprecipitation assays offer high sensitivity and specificity, enabling the detailed analysis of epigenetic changes in clinical samples. These tools are instrumental in translating epigenetic research into clinical practice. This review underscores the potential of epigenetic biomarkers to enhance the early diagnosis and prognosis of AD, paving the way for personalized therapeutic strategies and improved patient outcomes. The integration of these biomarkers into clinical workflows promises to revolutionize AD management, offering hope for better disease monitoring and intervention.
Collapse
Affiliation(s)
- Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab 140306, India.
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University, Jaipur, India
| | - Renu Arya
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Mohammad Khalid
- Department of pharmacognosy, College of pharmacy, Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Monika Sachdeva
- Fatima College of Health Sciences, Al Ain, United Arab Emirates
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box No. 71666, Riyadh 11597, Saudi Arabia
| | - Pratap Kumar Patra
- School of Pharmacy & Life Sciences, Centurion University of Technology & Managemnet, Bhubaneswar, Odisha 752050, India
| | - Ankur Agrawal
- Jai Institute of Pharmaceutical Sciences and Research, Gwalior, Madhya Pradesh 474001, India
| | - Pranay Wal
- PSIT-Pranveer Singh Institute of Technology, Pharmacy, NH-19, Bhauti Road, Kanpur, UP 209305, India
| | - Amin Gasmi
- Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France; International Institute of Nutrition and Micronutrition Sciences, Saint-Étienne, France
| |
Collapse
|
50
|
Gu YY, Zhao XR, Zhang N, Yang Y, Yi Y, Shao QH, Liu MX, Zhang XL. Mitochondrial dysfunction as a therapeutic strategy for neurodegenerative diseases: Current insights and future directions. Ageing Res Rev 2024; 102:102577. [PMID: 39528070 DOI: 10.1016/j.arr.2024.102577] [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: 09/19/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative diseases, as common diseases in the elderly, tend to become younger due to environmental changes, social development and other factors. They are mainly characterized by progressive loss or dysfunction of neurons in the central or peripheral nervous system, and common diseases include Parkinson's disease, Alzheimer's disease, Huntington's disease and so on. Mitochondria are important organelles for adenosine triphosphate (ATP) production in the brain. In recent years, a large amount of evidence has shown that mitochondrial dysfunction plays a direct role in neurodegenerative diseases, which is expected to provide new ideas for the treatment of related diseases. This review will summarize the main mechanisms of mitochondrial dysfunction in neurodegenerative diseases, as well as collating recent advances in the study of mitochondrial disorders and new therapies.
Collapse
Affiliation(s)
- Ying-Ying Gu
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Xin-Ru Zhao
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Nan Zhang
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Yuan Yang
- Department of Gastroenterology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Ying Yi
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Qian-Hang Shao
- Department of Pharmacy, Peking University People's Hospital, Beijing 100871, P R China
| | - Ming-Xuan Liu
- College of Pharmacy, Nantong University, Nantong 226001, PR China.
| | - Xiao-Ling Zhang
- College of Pharmacy, Nantong University, Nantong 226001, PR China.
| |
Collapse
|