1
|
Johnson CN, Evans MR, Blankenship AE, John CS, Rekowski MJ, Washburn MP, Phan A, Gouvion CM, Haeri M, Swerdlow RH, Geiger PC, Morris JK. Human skeletal muscle mitochondrial pathways are impacted by a neuropathologic diagnosis of Alzheimer's disease. Neurobiol Dis 2025; 210:106916. [PMID: 40250718 DOI: 10.1016/j.nbd.2025.106916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Alzheimer's disease (AD) is associated with reduced lean mass and impaired skeletal muscle mitochondrial and motor function. Although primary mitochondrial defects in AD may underlie these findings, molecular alterations in AD have not been thoroughly examined in human skeletal muscle. Here, we used two human skeletal muscle types, quadriceps (n = 81) and temporalis (n = 66), to compare the proteome of individuals with a neuropathologic AD diagnosis based on AD Neuropathologic Change (ADNPC+: n = 54 temporalis, 44 quadriceps) to controls (ADNPC-: n = 27 temporalis, 22 quadriceps). We determined the effects of ADNPC status within each muscle and within apolipoprotein E4 (APOE4) carriers and APOE4 non-carriers. Pathways that support mitochondrial metabolism, including oxidative phosphorylation, were downregulated in skeletal muscle of ADNPC+ versus ADNPC- individuals. Similar mitochondrial effects were observed across muscle types and APOE4 carrier groups, but nearly four times as many proteins were altered in temporalis versus quadriceps tissue and mitochondrial effects were most pronounced in APOE4 carriers compared to APOE4 non-carriers. Of all detected oxidative phosphorylation proteins, the expression of ∼29-61 % (dependent on muscle/APOE4 carrier group) significantly correlated with AD progression, ranked by Clinical Dementia Rating and ADNPC scores. Of these, 23 proteins decreased in expression with greater AD progression in all skeletal muscle type and APOE4 carrier groups. This is the first study to assess differences in the human skeletal muscle proteome in the context of AD. Our work shows that systemic mitochondrial alterations in AD extend to skeletal muscle and these effects are amplified by APOE4 and correlate with AD progression.
Collapse
Affiliation(s)
- Chelsea N Johnson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Mara R Evans
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anneka E Blankenship
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - Casey S John
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Michaella J Rekowski
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Michael P Washburn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Andy Phan
- Bruker Daltonics, Inc, Billerica, MA 01821, USA.
| | - Cynthia M Gouvion
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Mohammad Haeri
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Paige C Geiger
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
2
|
Garcia TFM, Desio JAF, de Souza EF, Henkes SFC, Santos LS, de Carvalho Muenho J, Gonçlaves CL, Dos Santos JCC. The silent saboteur: oxidative stress and the path to cognitive dysfunction. Neurodegener Dis Manag 2025:1-28. [PMID: 40424201 DOI: 10.1080/17582024.2025.2510175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
Oxidative stress (OS) plays a central role in age-related cognitive decline and neurodegeneration and is increasingly recognized as a key factor in the pathogenesis of Alzheimer's disease (AD) and Parkinson's disease (PD). Elevated OS biomarkers are detectable from the earliest stages of these disorders. In this critical narrative review, we explore the bioenergetic cascade underlying neurodegeneration, emphasizing pathophysiological alterations, mechanisms, and therapeutic targets. Recent evidence suggests that OS and impaired cellular energy dynamics are both early markers and downstream effects of neuroinflammation, contributing to symptom severity and reduced treatment efficacy. A deeper understanding of these interrelated processes is essential for the development of more effective interventions. Monitoring OS-related metabolites may offer a promising strategy for identifying therapeutic targets and enabling early clinical intervention, ultimately aiming to reduce neuroinflammation and improve patient outcomes in AD and PD.
Collapse
Affiliation(s)
- Tulia Fernanda Meira Garcia
- Multicampi School of Medical Sciences, Federal University of Rio Grande do Norte, Caicó, Rio Grande do Norte, Brazil
| | | | | | | | - Luana Stangherlin Santos
- Department of Cardiology, Complexo Hospitalar de Doenças Cardiopulmonares (CDAN), Luanda, Angola
| | - Julcileia de Carvalho Muenho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, Santa Catarina, Angola
| | - Cinara Ludvig Gonçlaves
- Department of Cardiology, Complexo Hospitalar de Doenças Cardiopulmonares (CDAN), Luanda, Angola
| | | |
Collapse
|
3
|
Chakraborty S, Rao S, Tripathi SJ. The neuroprotective effects of N-acetylcysteine in psychiatric and neurodegenerative disorders: From modulation of glutamatergic transmission to restoration of synaptic plasticity. Neuropharmacology 2025:110527. [PMID: 40414419 DOI: 10.1016/j.neuropharm.2025.110527] [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: 03/01/2025] [Revised: 05/10/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
N-acetylcysteine (NAC) is an effective pleiotropic drug with a strong safety profile. It is predominantly used as a mucolytic agent and in the treatment of paracetamol overdose. However, extensive research in the last decade has shown the prominent efficacy of NAC in many neuropsychiatric and neurodegenerative disorders. NAC acts through multiple mechanisms; primarily, it releases cysteine and modulates glutamatergic and monoaminergic transmission. Further, it restores glutathione levels, promotes oxidative balance, reverses decreased synaptic plasticity, reduces neuroinflammation and mitochondrial dysfunction, and provides neurotrophic support. Additionally, it regulates one-carbon metabolism pathways, leading to the production of key metabolites. In this review, we will be discussing in-depth mechanisms of action of NAC and its promising ability to reverse neuropathological changes, particularly cognitive deficits, and associated plasticity changes in various psychiatric and neurodegenerative diseases, including depression, bipolar disorders, schizophrenia, Alzheimer's disease, Huntington's disease, traumatic brain injury, aging. Overall, several preclinical studies and clinical trials have demonstrated the efficacy of NAC in reversing regressive plasticity, cognitive deficits, and associated changes in the brain. NAC remains among the strongest candidates with a high safety profile for managing several types of neurological disorders.
Collapse
Affiliation(s)
- Suwarna Chakraborty
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shankaranarayana Rao
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India.
| | - Sunil Jamuna Tripathi
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
4
|
Jia SZ, Li Y, Xu XW, Huang YP, Deng XY, Zhang ZH, Song GL. Selenoprotein K Confers Protection against Iron Dyshomeostasis-Related Neurotoxicity by Regulating the Palmitoylation of TfR-1. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12233-12246. [PMID: 40296316 PMCID: PMC12100729 DOI: 10.1021/acs.jafc.4c08266] [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] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Selenoprotein K (SELENOK), a protein residing in the endoplasmic reticulum (ER), is modulated by dietary selenium and is expressed at elevated levels in neurons. SELENOK has been shown to participate in cellular antioxidant activity and posttranslational palmitoylation. This study presents both in vivo and in vitro evidence that SELENOK deficiency reduces the palmitoylation of TfR-1, thereby impairing transferrin transport and ultimately leading to a decrease in the intracellular iron content, impaired mitochondrial respiratory chain activity and decreased ATP production. Remarkably, restoring SELENOK levels significantly enhanced TfR-1 palmitoylation, increased intracellular iron levels, and restored mitochondrial function, thus ameliorating cognitive deficits in 7 month-old SELENOK knockout mice. Consistent with these findings, iron supplementation also improved mitochondrial function by elevating intracellular iron levels, thereby improving cognitive deficits in 7 month-old SELENOK knockout mice. Therefore, SELENOK exerts its neuroprotective effect by regulating the palmitoylation of TfR-1 to maintain iron homeostasis, thereby protecting mitochondrial function in neurons.
Collapse
Affiliation(s)
- Shi-Zheng Jia
- Shenzhen
Key Laboratory of Marine Bioresources and Ecology, Brain Disease and
Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen518060, China
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Shenzhen
University Health Science Center, Shenzhen518060, China
| | - Yu Li
- Shenzhen
Key Laboratory of Marine Bioresources and Ecology, Brain Disease and
Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen518060, China
| | - Xin-Wen Xu
- Shenzhen
Key Laboratory of Marine Bioresources and Ecology, Brain Disease and
Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen518060, China
| | - Yan-Ping Huang
- Shenzhen
Key Laboratory of Marine Bioresources and Ecology, Brain Disease and
Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen518060, China
| | - Xiao-Yi Deng
- Shenzhen
Key Laboratory of Marine Bioresources and Ecology, Brain Disease and
Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen518060, China
| | - Zhong-Hao Zhang
- Shenzhen
Key Laboratory of Marine Bioresources and Ecology, Brain Disease and
Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen518060, China
- Shenzhen-Hong
Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen518060, China
| | - Guo-Li Song
- Shenzhen
Key Laboratory of Marine Bioresources and Ecology, Brain Disease and
Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen518060, China
- Shenzhen-Hong
Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen518060, China
| |
Collapse
|
5
|
Li J, Liu T, Xian M, Wei J. Therapeutic applications of exercise in neurodegenerative diseases: focusing on the mechanism of SIRT1. Mol Cell Biochem 2025:10.1007/s11010-025-05299-8. [PMID: 40358811 DOI: 10.1007/s11010-025-05299-8] [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: 02/06/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
Neurodegenerative diseases comprise a group of central nervous system disorders marked by progressive neuronal degeneration and dysfunction. Their pathogenesis is multifactorial, involving oxidative stress, mitochondrial dysfunction, excitotoxicity, and neuroinflammation. Recent research has highlighted the potential of exercise as a non-pharmacological intervention for both the prevention and treatment of these disorders. In particular, exercise has received growing attention for its capacity to upregulate the expression and activity of SIRT1, a critical mediator of neuroprotection via downstream signaling pathways. SIRT1, a key member of the Sirtuin family, is a nicotinamide adenine dinucleotide (NAD +)-dependent class III histone deacetylase. It plays an essential role in regulating cellular metabolism, energy homeostasis, gene expression, and cellular longevity. In the context of neurodegenerative diseases, SIRT1 confers neuroprotection by modulating multiple signaling cascades through deacetylation, suppressing neuronal apoptosis, and promoting neural repair and regeneration. Exercise enhances SIRT1 expression and activity by increasing NAD + synthesis and utilization, improving intracellular redox balance, alleviating oxidative stress-induced inhibition of SIRT1, and thereby promoting its activation. Moreover, exercise may indirectly modulate SIRT1 function by influencing interacting molecular networks. This review summarizes recent advances in the therapeutic application of exercise for neurodegenerative diseases, with a focus on SIRT1 as a central mechanism. It examines how exercise mediates neuroprotection through the regulation of SIRT1 and its associated molecular mechanisms and signaling pathways. Finally, the paper discusses the potential applications and challenges of integrating exercise and SIRT1-targeted strategies in the management of neurodegenerative diseases, offering novel perspectives for the development of innovative treatments and improvements in patients' quality of life.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, 475004, Henan, China
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, 475004, Henan, China.
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
6
|
Smith LA, Keane EB, Connor K, Chan F, Cunningham MO. In vitro modelling of the neuropathophysiological features of mitochondrial epilepsy. Seizure 2025:S1059-1311(25)00121-9. [PMID: 40410091 DOI: 10.1016/j.seizure.2025.05.005] [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: 04/09/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/25/2025] Open
Abstract
Epilepsy is a common and severe neurological manifestation of primary mitochondrial disease, affecting approximately 60 % of paediatric patients and 20 % of adult patients. Many of the mitochondrial epilepsies, particularly those presenting in childhood, are refractory to anti-epileptic treatment. Moreover, these conditions are typically characterised by severe neurodegeneration and closely associated with neurological decline and premature death. Indeed, there persists an urgent need to delineate the mechanisms underpinning mitochondrial epilepsy in order to develop effective treatments. In this review, we provide an overview of currently available in vitro models of the mitochondrial epilepsies. Such models offer opportunities to characterise early disease pathophysiology and interrogate novel mitochondrial-targeting and anti-epileptic treatments, with an overall aim to modulate seizure associated pathology and activity for the mitochondrial epilepsies. We discuss the use of acute cortical and subcortical brain slice preparations, obtained from both neurosurgical patients and rodents, for modelling the common neuropathophysiological features of mitochondrial epilepsy. We also review the use of induced pluripotent stem cell derived neural and glial culture models, and the development of three-dimensional cerebral organoids, generated from fibroblasts obtained from patients with primary mitochondrial disease. Human-derived, disease-relevant in vitro model systems which recapitulate the complexity and pathological features observed in patient brain tissues are crucial to help bridge the gap between animal models and patients living with mitochondrial epilepsy.
Collapse
Affiliation(s)
- Laura A Smith
- Mitochondrial Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - Ella B Keane
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Ireland
| | - Kate Connor
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Ireland
| | - Felix Chan
- Department of Pharmacy, School of Health Sciences, University of Birmingham, Birmingham, B15 2TT, UK; Centre for Human Brain Health (CHBH), University of Birmingham, Birmingham, B15 2TT, UK; Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, B15 2TT, UK
| | - Mark O Cunningham
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Ireland.
| |
Collapse
|
7
|
Hu Y, Liu Y, Zhu Q, Chen Y, Zeng Y. Identification of Novel Biomarkers Related to Vesicle Trafficking in Alzheimer's Disease Using Bioinformatics Approaches. Neurochem Res 2025; 50:157. [PMID: 40338387 DOI: 10.1007/s11064-025-04410-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with complex pathogenesis. Vesicle trafficking abnormalities are closely associated with AD, making the identification of related biomarkers crucial. Chip data of AD were downloaded from the GEO database as training and test sets. Differentially expressed vesicle trafficking-related genes were analyzed, followed by construction of protein-protein interaction (PPI) networks, machine learning for important biomarkers identification, and various analyses including ROC curve analysis, and construction of regulatory networks. A total of 149 differentially expressed vesicle trafficking-related genes were identified. Through multiple analyses, 5 key genes (KIF22, ACTR10, TUBB2A, TUBA3C, and DCTN1) were obtained. Additionally, potential miRNA regulatory networks and candidate drugs were predicted, and AD subtypes were characterized.This study successfully identified novel biomarkers related to vesicle trafficking in AD, and these findings provide new insights into the role of intracellular transport dysfunction in AD pathogenesis.
Collapse
Affiliation(s)
- Yirong Hu
- Department of Neurology, Yichun People's Hospital, No. 1061 Jinxiu Avenue, Yiyang New District, Yichun, Jiangxi, 336000, China
| | - Yi Liu
- Cardiothoracic surgery, Yichun People's Hospital, Yichun, Jiangxi, 336000, China
| | - Qiuyan Zhu
- Department of Neurology, Yichun People's Hospital, No. 1061 Jinxiu Avenue, Yiyang New District, Yichun, Jiangxi, 336000, China
| | - Yong Chen
- Department of Neurology, Yichun People's Hospital, No. 1061 Jinxiu Avenue, Yiyang New District, Yichun, Jiangxi, 336000, China
| | - Ying Zeng
- Department of Neurology, Yichun People's Hospital, No. 1061 Jinxiu Avenue, Yiyang New District, Yichun, Jiangxi, 336000, China.
| |
Collapse
|
8
|
Lei S, Liu Y. Identifying blood mitochondrial DNA copy number as a biomarker for development of neurodegenerative diseases: Evidence from Mendelian randomization analysis. Neuroscience 2025; 573:421-429. [PMID: 40185386 DOI: 10.1016/j.neuroscience.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Mitochondrial dysfunction has been associated with neurodegenerative diseases (NDDs). This study aimed to explore the association between blood mitochondrial DNA copy number (mtDNA-CN) and development of NDDs. This study was based on two-sample Mendelian randomization (MR) analysis. The genome wide association study (GWAS) data of NDDs including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), age-related macular degeneration (AMD), multiple sclerosis (MS), Parkinson's disease (PD), primary open-angle glaucoma (POAG), and vascular dementia (VD) was obtained from FinnGen consortium. Inverse-variance weighted (IVW) was applied as the primary approach for MR estimation. MR results revealed that blood mtDNA-CN exhibited a significant relationship with the incidence of AD (IVW-P = 0.011, odds ratio [OR] = 0.65) and AMD (IVW-P = 0.038, OR = 0.64). However, there was no significant association observed between blood mtDNA-CN and other NDDs (IVW-P > 0.05). Our findings supported the relationship between mitochondrial dysfunction and development of AD and AMD, and that blood mtDNA-CN may serve as a potential biomarker for the incidence of these two NDDs.
Collapse
Affiliation(s)
- Shizhen Lei
- Department of Ophthalmology, Wuhan No.1 Hospital, Wuhan, Hubei, China.
| | - Yani Liu
- Department of Otolaryngology & Head and Neck Surgery, Wuhan No.1 Hospital, Wuhan, Hubei, China
| |
Collapse
|
9
|
Jin J, Yue L, Du M, Geng F, Gao X, Zhou Y, Lu Q, Pan X. Molecular Hydrogen Therapy: Mechanisms, Delivery Methods, Preventive, and Therapeutic Application. MedComm (Beijing) 2025; 6:e70194. [PMID: 40297245 PMCID: PMC12035766 DOI: 10.1002/mco2.70194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Molecular hydrogen (H2), recognized as the smallest gas molecule, is capable of permeating cellular membranes and diffusing throughout the body. Due to its high bioavailability, H2 is considered a therapeutic gas for the treatment of various diseases. The therapeutic efficacy of hydrogen is contingent upon factors such as the administration method, duration of contact with diseased tissue, and concentration at targeted sites. H2 can be administered exogenously and is also produced endogenously within the intestinal tract. A comprehensive understanding of its delivery mechanisms and modes of action is crucial for advancing hydrogen medicine. This review highlights H₂'s mechanisms of action, summarizes its administration methods, and explores advancements in treating intestinal diseases (e.g., inflammatory bowel disease, intestinal ischemia-reperfusion, colorectal cancer). Additionally, its applications in managing other diseases are discussed. Finally, the challenges associated with its clinical application and potential solutions are explored. We propose that current delivery challenges faced by H2 can be effectively addressed through the use of nanoplatforms; furthermore, interactions between hydrogen and gut microbiota may provide insights into its mechanisms for treating intestinal diseases. Future research should explore the synergistic effects of H2 in conjunction with conventional therapies and develop personalized treatment plans to achieve precision medicine.
Collapse
Affiliation(s)
- Jiayi Jin
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Lijun Yue
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Maoru Du
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Feng Geng
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Xue Gao
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Yuming Zhou
- Department of Laboratory MedicineYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Qianqian Lu
- Department of OncologyYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Xiaohong Pan
- School of PharmacyBinzhou Medical UniversityYantaiChina
| |
Collapse
|
10
|
Lei S, Liu Y. Identifying microglia-derived NFKBIA as a potential contributor to the pathogenesis of Alzheimer's disease and age-related macular degeneration. J Alzheimers Dis 2025; 105:134-146. [PMID: 40105475 DOI: 10.1177/13872877251326267] [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] [Indexed: 03/20/2025]
Abstract
BackgroundAlzheimer's disease (AD) and age-related macular degeneration (AMD) place considerable health burden on affected individuals and significant economic burden on society.ObjectiveThis study aims to explore the shared cellular and molecular mechanisms underlying the pathogenesis of AD and AMD.MethodsThe investigation in this study is conducted via single-cell and bulk tissue transcriptomic analysis. Transcriptomic datasets of AD and AMD were obtained from the GEO database. The shared differentially expressed genes (DEGs) in control and AD- and AMD-affected samples were identified. Functional enrichment analysis for DEGs was subsequently performed. Then, the protein-protein interaction (PPI) network of these DEGs was established via the STRING database and hub genes of this network were identified by Cytoscape software. Single-cell transcriptomic analysis was performed using Seurat R package to explore their expression in different cell types.ResultsDifferential analysis identified 127 shared DEGs of the two diseases, including 71 upregulated and 56 downregulated genes. Upregulated DEGs were enriched in inflammation, gliogenesis, cell apoptosis, and response to bacterial and viral infection and downregulated DEGs were enriched in mitochondrial function and energy production. PPI network and Cytoscape determined 10 hub genes, of which the NFKBIA gene was associated with the severity of both AD and AMD. Moreover, single-cell transcriptomic analysis showed that NFKBIA was highly expressed in microglia from disease-affected tissues.ConclusionsThe findings indicated that microglia with high NFKBIA expression were important contributors to the progression of both AD and AMD. Microglia-derived NFKBIA might serve as a potential therapeutic target for AD and AMD.
Collapse
Affiliation(s)
- Shizhen Lei
- Department of Ophthalmology, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Yani Liu
- Department of Otolaryngology & Head and Neck Surgery, Wuhan No.1 Hospital, Wuhan, Hubei, China
| |
Collapse
|
11
|
Yan B, Zhou J, Yan F, Gao M, Tang J, Huang L, Luo Y. Unlocking the potential of photobiomodulation therapy for brain neurovascular coupling: The biological effects and medical applications. J Cereb Blood Flow Metab 2025; 45:800-830. [PMID: 39763390 PMCID: PMC11705326 DOI: 10.1177/0271678x241311695] [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: 06/04/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025]
Abstract
Photobiomodulation (PBM) therapy stands as an innovative neurostimulation modality that has demonstrated both efficacy and safety in improving brain function. This therapy exerts multifaceted influences on neurons, blood vessels, and their intricate interplay known as neurovascular coupling (NVC). Growing evidence indicates that NVC may present a promising target for PBM intervention. However, the detailed mechanisms underlying its therapeutic benefits remain to be fully understood. This review aims to elucidate the potential metabolic pathways and signaling cascades involved in the modulatory effects of PBM, while also exploring the extensive repertoire of PBM applications in neurologic and psychiatric conditions. The prospects of PBM within the realm of NVC investigation are intensively considered, providing deeper insights into the powerful capabilities of PBM therapy and its potential to revolutionize neurostimulation treatments.
Collapse
Affiliation(s)
- Bingzi Yan
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhou
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Fengshuo Yan
- The Second Research Institute of Civil Aviation Administration of China, Chengdu, China
| | - Mingyang Gao
- The Second Research Institute of Civil Aviation Administration of China, Chengdu, China
| | - Jiaji Tang
- Sichuan Becoming Technology Co., LTD, Chengdu, China
| | - Lin Huang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Luo
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Kärkkäinen V, Saari T, Hannonen S, Rusanen M, Lehtola JM, Uusitalo H, Leinonen V, Thiede B, Kaarniranta K, Koivisto AM, Utheim TP. Altered tear fluid protein expression in persons with mild Alzheimer's disease in proteins involved in oxidative stress, protein synthesis, and energy metabolism. J Alzheimers Dis 2025; 105:292-301. [PMID: 40183343 DOI: 10.1177/13872877251326868] [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] [Indexed: 04/05/2025]
Abstract
BackgroundTear fluid (TF) is a protein-rich solution that reflects pathophysiological changes in Alzheimer's disease (AD).ObjectiveIn this study, we examined whether TF proteins were differently expressed in persons with mild AD dementia compared to cognitively healthy controls (CO).MethodsWe analyzed data from 53 study participants including 34 CO (mean age, 71 years; Mini-Mental State Examination [MMSE] score, 28.9 ± 1.4), and 19 patients with AD (Clinical Dementia Rating, 0.5-1; mean age, 72 years; MMSE score, 23.8 ± 2.8). All participants underwent cognitive testing, as well as neurological and ophthalmological examinations. TF was collected using Schirmer strips, and TF protein content was evaluated using mass spectrometry-based proteomics and label-free quantification.ResultsWe found that 16 proteins exhibited significantly upregulated expression in the AD group compared to the CO group (p ≤ 0.05). These proteins were NP1L4, BBOX1, CYTC, RNAS4, PCD, RNT2, AL1A3, SYSC, TPIS, CLH1, PGAM1, EIF3L, 5NTC, HNRNPA2B1, PYGL, and ERO1α. No proteins were significantly downregulated in the AD group compared to the CO group.ConclusionsOur results support the hypothesis that TF is a potential source of biomarkers for AD. Part of those proteins with altered expression have previously linked to increased oxidative stress, changed protein synthesis, and disturbed regulation of energy metabolism related to AD or neurodegenerative disease. The present results indicate the value of continued investigation of TF proteins in AD.
Collapse
Affiliation(s)
- Virve Kärkkäinen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- NeuroCenter, Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Toni Saari
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sanna Hannonen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Minna Rusanen
- Ceriatric Center, Wellbeing Services Country of North Karelia, Joensuu, Finland
| | - Juha-Matti Lehtola
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Hannu Uusitalo
- Eye and Vision Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ville Leinonen
- NeuroCenter, Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Anne M Koivisto
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Department of Geriatrics, Helsinki University Hospital and Department of Neurosciences, University of Helsinki, Helsinki, Finland
| | - Tor P Utheim
- Department of Ophthalmology, University of Oslo, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
Dölle C, Tzoulis C. NAD augmentation as a disease-modifying strategy for neurodegeneration. Trends Endocrinol Metab 2025:S1043-2760(25)00070-0. [PMID: 40287324 DOI: 10.1016/j.tem.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Neurodegenerative diseases (NDDs) pose a significant and rapidly growing global health challenge, but there are no effective therapies to delay or halt progression. In recent years augmentation of nicotinamide adenine dinucleotide (NAD) has emerged as a promising disease-modifying strategy that targets multiple key disease pathways across multiple NDDs, such as mitochondrial dysfunction, energy deficits, proteostasis, and neuroinflammation. Several early clinical trials of NAD augmentation have been completed, and many more are currently underway, reflecting the growing optimism and urgency within the field. We discuss the rationale and evolving therapeutic landscape of NAD augmentation. We argue that, to fully realize its therapeutic potential, it is essential to determine the specific contexts in which NAD supplementation is most effective and to address crucial knowledge gaps.
Collapse
Affiliation(s)
- Christian Dölle
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, 5020 Bergen, Norway; K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Pb 7804, 5020 Bergen, Norway.
| | - Charalampos Tzoulis
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, 5020 Bergen, Norway; K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Pb 7804, 5020 Bergen, Norway.
| |
Collapse
|
14
|
Khan B, Iqbal MK, Khan MA, Khan H, Kiyani MM, Bashir S, Li S. Unraveling the Complexity of Alzheimer's Disease: Insights into Etiology and Advancements in Treatment Strategies. J Mol Neurosci 2025; 75:57. [PMID: 40279003 DOI: 10.1007/s12031-025-02337-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/20/2025] [Indexed: 04/26/2025]
Abstract
Alzheimer's disease, a complex and progressive neurological disorder, is the leading cause of late-life dementia. Pathologically, it is marked by the presence of amyloid plaques and neurofibrillary tangles in the brain. Over the past two decades, advancements in understanding the disease's pathogenesis have spurred research into new pharmacological treatments that target its underlying mechanisms. Currently available drugs, such as acetylcholinesterase inhibitors (rivastigmine, galantamine, donepezil) and the NMDA receptor antagonist memantine, primarily address symptoms and are effective only in the later stages of the disease. While these medications can slow disease progression and provide symptomatic relief, they do not offer a cure. Despite having a clear understanding of Alzheimer's neuropathology, the precise mechanisms driving the disease remain elusive. The lack of effective treatments that can stop the start and progression of the disease may be caused by our incomplete understanding of the pathogenic process. New therapeutic targets are now available due to the significant advancements made in pathophysiology over the past few years, which should allow for a direct attack on the underlying illness process. The various pathophysiological pathways that underlie Alzheimer's disease and how it is managed by conventional medication therapy, including current exploratory therapeutic options, are covered in this review article. Innovative, beneficial policies are essential to determine and progress therapeutic molecules to defend against AD.
Collapse
Affiliation(s)
- Bakhtawar Khan
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Muhammad Khalid Iqbal
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Muhammad Ajmal Khan
- Division of Life Sciences, Center for Cancer Researchand, State Key Laboratory of Molecular Neurosciencesaq, The Hong Kong University of Science and Technology, Clear Water Bay Hong Kong, China
| | - Hamid Khan
- Department of Biological Science, International Islamic University, Islamabad, Pakistan
| | - Mubin Mustafa Kiyani
- Shifa College of Medical Technology, Shifa Tameer-E-Millat University, Islamabad, Pakistan
| | - Shahid Bashir
- Department of Neuroscience, King Fahad Hospital, Dammam, Saudi Arabia.
- King Salman Center for Disability Research, 11614, Riyadh, Saudi Arabia.
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China.
| |
Collapse
|
15
|
Zhang S, Zhu M, Chen S. Exploring the Interconnections Between Mitochondrial Dysfunction and Polycystic Ovary Syndrome: A Comprehensive Integrated Analysis. Biochem Genet 2025:10.1007/s10528-025-11104-4. [PMID: 40259200 DOI: 10.1007/s10528-025-11104-4] [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: 11/08/2024] [Accepted: 04/09/2025] [Indexed: 04/23/2025]
Abstract
Polycystic ovary syndrome (PCOS) is a leading cause of anovulatory infertility and is strongly linked to mitochondrial dysfunction (MD) in reproductive-age women. MD contributes to excessive reactive oxygen species (ROS) accumulation, exacerbating disease progression. This study aimed to identify key MD-related genes (MDRGs) involved in PCOS through bioinformatics analyses and experimental validation. Two PCOS transcriptome datasets (GSE34526 and GSE5850) were analyzed to identify differentially expressed genes (DEGs), which were then intersected with MDRGs to obtain MD-related DEGs (MDDEGs). Functional enrichment (GO, KEGG, GSEA) and protein-protein interaction (PPI) network analyses identified eight hub MDDEGs (MMP9, PPP1 CA, PSMD12, LIFR, PRKAA1, ITGAM, SUCLA2, GPBAR1). A rat PCOS model was established to validate hub gene expression via RT-qPCR, western blotting, and immunohistochemistry. The experimental data confirmed that seven hub genes exhibited consistent expression patterns with GSE34526 (P < 0.05), while only PRKAA1 and LIFR matched GSE5850 findings. Additionally, ROC analysis for the five most significant genes (LIFR, PBK, PRKAA1, RCAN1, MMP9) demonstrated promising diagnostic value (AUC > 0.85). This study highlights the role of MD in shaping the immune microenvironment of PCOS and identifies novel molecular targets for potential therapeutic interventions.
Collapse
Affiliation(s)
- Suqin Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China
| | - Mingyue Zhu
- Department of Gynecology and Obstetrics Zhujiang Hospital, Southern Medical University, No.253 Guangzhou Industrial Avenue Road, Guangzhou, 510515, Guangdong, China
| | - Shiling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics Nanfang Hospital, Southern Medical University, No.1838 Guangzhou Northern Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
16
|
刘 露, 公 茂, 廖 国, 赵 维, 傅 强. [Hypertension exacerbates postoperative learning and memory impairment in rats possibly due to UCP2 downregulation-mediated mitochondrial dysfunction]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:725-735. [PMID: 40294922 PMCID: PMC12037294 DOI: 10.12122/j.issn.1673-4254.2025.04.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Indexed: 04/30/2025]
Abstract
OBJECTIVES To explore the correlation of hypertension with postoperative cognitive dysfunction and its possible mechanism. METHODS Twelve-week-old spontaneously hypertensive rats (SHRs) and Wistar-Kyoto (WKY) rats were both randomized into control group and surgical group (n=8). In the latter group, the rats received carotid artery exposure surgery under sevoflurane anesthesia to establish models of postoperative learning and memory impairment. Postoperative cognitive function changes of the rats were evaluated using behavioral tests. The hippocampus of the rats were collected for determining ATP level and mitochondrial membrane potential (MMP) and for detecting expressions of UCP2 and astrocyte markers (GFAP and NOX4) using Western blotting and immunofluorescence staining. Serum levels of ROS, IL-6, IL-1β and TNF‑α were detected using ELISA. Nissl staining was used to examine hippocampal neuronal loss in the CA1 region. RESULTS The SHRs exhibited exacerbated learning and memory deficits following the surgery as shown by significantly reduced performance in novel object recognition tests and context-related and tone-related fear conditioning experiments. Compared with WKY rats, the SHRs had significantly decreased mitochondrial UCP2 expression and MMP in the hippocampus, increased hippocampal ATP level, and markedly increased serum levels of ROS and inflammatory factors, showing also increased activation of hippocampal astrocytes and microglia and reduced number of neurons positive for Nissl staining. CONCLUSIONS Hypertension can exacerbate major postoperative learning and memory impairment in rats possibly as a result of UCP2-mediated mitochondrial dysfunction and oxidative stress damage, which further leads to astrocyte overactivation and neuronal damage.
Collapse
Affiliation(s)
| | | | | | | | - 强 傅
- 傅 强,博士生导师,教授,主任医师,E-mail:
| |
Collapse
|
17
|
Zhao H, Qiu X, Wang S, Wang Y, Xie L, Xia X, Li W. Multiple pathways through which the gut microbiota regulates neuronal mitochondria constitute another possible direction for depression. Front Microbiol 2025; 16:1578155. [PMID: 40313405 PMCID: PMC12043685 DOI: 10.3389/fmicb.2025.1578155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
As a significant mental health disorder worldwide, the treatment of depression has long faced the challenges of a low treatment rate, significant drug side effects and a high relapse rate. Recent studies have revealed that the gut microbiota and neuronal mitochondrial dysfunction play central roles in the pathogenesis of depression: the gut microbiota influences the course of depression through multiple pathways, including immune regulation, HPA axis modulation and neurotransmitter metabolism. Mitochondrial function serves as a key hub that mediates mood disorders through mechanisms such as defective energy metabolism, impaired neuroplasticity and amplified neuroinflammation. Notably, a bidirectional regulatory network exists between the gut microbiota and mitochondria: the flora metabolite butyrate enhances mitochondrial biosynthesis through activation of the AMPK-PGC1α pathway, whereas reactive oxygen species produced by mitochondria counteract the flora composition by altering the intestinal epithelial microenvironment. In this study, we systematically revealed the potential pathways by which the gut microbiota improves neuronal mitochondrial function by regulating neurotransmitter synthesis, mitochondrial autophagy, and oxidative stress homeostasis and proposed the integration of probiotic supplementation, dietary fiber intervention, and fecal microbial transplantation to remodel the flora-mitochondrial axis, which provides a theoretical basis for the development of novel antidepressant therapies targeting gut-brain interactions.
Collapse
Affiliation(s)
- Hongyi Zhao
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiongfeng Qiu
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuyu Wang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Wang
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Xie
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiuwen Xia
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weihong Li
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan College of Traditional Chinese Medicine, Mianyang, China
| |
Collapse
|
18
|
Jellinger KA. Concomitant Pathologies and Their Impact on Parkinson Disease: A Narrative Overview of Current Evidence. Int J Mol Sci 2025; 26:2942. [PMID: 40243562 PMCID: PMC11988849 DOI: 10.3390/ijms26072942] [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: 02/26/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Many clinico-pathological studies point to the presence of multiple comorbidities/co-pathologies in the course of Parkinson disease (PD). Lewy body pathology, the morphological hallmark of PD, rarely exists in isolation, but is usually associated with other concomitant pathologies, in particular Alzheimer disease-related changes (ADNC), cerebrovascular pathologies (macro- and microinfarcts, cerebral small vessel disease, cerebral amyloid angiopathy), TDP-43 pathology as well as multiple pathological combinations. These include cardiovascular disorders, metabolic syndrome, diabetes mellitus, autoimmune and rheumatic diseases, myasthenia gravis, Sjögren's syndrome, restless leg syndrome or other rare disorders, like Fabry disease. A combination of PD and multiple sclerosis (MS) may be due to the immune function of LRRK2 and its interrelation with α-synuclein. COVID-19 and HIV posed considerable impacts on patients with PD. Epidemiological evidence points to a decreased risk for the majority of neoplasms, except melanoma and other skin cancers, while some tumors (breast, brain) are increased. On the other hand, a lower frequency of malignancies preceding early PD markers may argue for their protective effect on PD risk. Possible pathogenetic factors for the association between PD and cancer are discussed. The tremendous heterogeneity of concomitant pathologies and comorbidities observed across the PD spectrum is most likely caused by the complex interplay between genetic, pathogenic and other risk factors, and further research should provide increasing insight into their relationship with idiopathic PD (and other parkinsonian disorders) in order to find better diagnostic tools and probable disease-modifying therapies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| |
Collapse
|
19
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. The Janus Face of Oxidative Stress and Hydrogen Sulfide: Insights into Neurodegenerative Disease Pathogenesis. Antioxidants (Basel) 2025; 14:360. [PMID: 40227410 PMCID: PMC11939184 DOI: 10.3390/antiox14030360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Oxidative stress plays an essential role in neurodegenerative pathophysiology, acting as both a critical signaling mediator and a driver of neuronal damage. Hydrogen sulfide (H2S), a versatile gasotransmitter, exhibits a similarly "Janus-faced" nature, acting as a potent antioxidant and cytoprotective molecule at physiological concentrations, but becoming detrimental when dysregulated. This review explores the dual roles of oxidative stress and H2S in normal cellular physiology and pathophysiology, focusing on neurodegenerative disease progression. We highlight potential therapeutic opportunities for targeting redox and sulfur-based signaling systems in neurodegenerative diseases by elucidating the intricate balance between these opposing forces.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
20
|
Aydın Ş, Özdemir S, Adıgüzel A. The Potential of cfDNA as Biomarker: Opportunities and Challenges for Neurodegenerative Diseases. J Mol Neurosci 2025; 75:34. [PMID: 40080233 PMCID: PMC11906534 DOI: 10.1007/s12031-025-02317-8] [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: 01/02/2025] [Accepted: 02/06/2025] [Indexed: 03/15/2025]
Abstract
Neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), are characterized by the progressive and gradual degeneration of neurons. The prevalence and rates of these disorders rise significantly with age. As life spans continue to increase in many countries, the number of cases is expected to grow in the foreseeable future. Early and precise diagnosis, along with appropriate surveillance, continues to pose a challenge. The high heterogeneity of neurodegenerative diseases calls for more accurate and definitive biomarkers to improve clinical therapy. Cell-free DNA (cfDNA), including fragmented DNA released into bodily fluids via apoptosis, necrosis, or active secretion, has emerged as a promising non-invasive diagnostic tool for various disorders including neurodegenerative diseases. cfDNA can serve as an indicator of ongoing cellular damage and mortality, including neuronal loss, and may provide valuable insights into disease processes, progression, and therapeutic responses. This review will first cover the key aspects of cfDNA and then examine recent advances in its potential use as a biomarker for neurodegenerative disorders.
Collapse
Affiliation(s)
- Şeyma Aydın
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Ahmet Adıgüzel
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
21
|
Delic S, Miletic Drakulic S, Stepovic M, Milosavljevic J, Kovacevic Dimitrijevic M, Jovanovic K, Marinkovic I, Tepavcevic M, Janicijevic N, Mitrovic A, Igrutinovic D, Vulovic M. The Connection Between Oxidative Stress, Mitochondrial Dysfunction, Iron Metabolism and Microglia in Multiple Sclerosis: A Narrative Review. NEUROSCI 2025; 6:23. [PMID: 40137866 PMCID: PMC11944927 DOI: 10.3390/neurosci6010023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
In recent years, in the pathogenesis of multiple sclerosis, emphasis has been placed on mitochondrial processes that influence the onset of the disease. Oxidative stress would be one of the consequences of mitochondrial dysfunction, and its impact on brain tissue is well described. Microglia, as a brain macrophage, have an important function in removing unwanted metabolites, as well as iron, which is an amplifier of oxidative stress. There are novelties in terms of the connection between these processes, which have redirected research more towards the process of neurodegeneration itself, so that the emphasis is no longer on neuroinflammation, which would initiate the pathological process itself and still exist in the vicinity of lesions with reduced intensity. The aim of this review is to summarize the current knowledge from the literature regarding oxidative stress, mitochondrial dysfunction and iron metabolism and how microglia are involved in these processes in multiple sclerosis.
Collapse
Affiliation(s)
- Simonida Delic
- Department of Anatomy, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.S.); (J.M.); (M.K.D.); (K.J.); (I.M.); (M.T.); (M.V.)
| | - Svetlana Miletic Drakulic
- Department of Neurology, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Milos Stepovic
- Department of Anatomy, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.S.); (J.M.); (M.K.D.); (K.J.); (I.M.); (M.T.); (M.V.)
| | - Jovana Milosavljevic
- Department of Anatomy, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.S.); (J.M.); (M.K.D.); (K.J.); (I.M.); (M.T.); (M.V.)
| | - Marija Kovacevic Dimitrijevic
- Department of Anatomy, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.S.); (J.M.); (M.K.D.); (K.J.); (I.M.); (M.T.); (M.V.)
| | - Kristijan Jovanovic
- Department of Anatomy, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.S.); (J.M.); (M.K.D.); (K.J.); (I.M.); (M.T.); (M.V.)
| | - Ivona Marinkovic
- Department of Anatomy, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.S.); (J.M.); (M.K.D.); (K.J.); (I.M.); (M.T.); (M.V.)
| | - Melanija Tepavcevic
- Department of Anatomy, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.S.); (J.M.); (M.K.D.); (K.J.); (I.M.); (M.T.); (M.V.)
| | - Nikoleta Janicijevic
- Department of Hygiene and Ecology, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Aleksandra Mitrovic
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Danica Igrutinovic
- Department of Biochemistry, University Clinical Centre Kragujevac, 34000 Kragujevac, Serbia;
| | - Maja Vulovic
- Department of Anatomy, Faculty of Medical Sciences Kragujevac, University of Kragujevac, 34000 Kragujevac, Serbia; (M.S.); (J.M.); (M.K.D.); (K.J.); (I.M.); (M.T.); (M.V.)
| |
Collapse
|
22
|
Liu H, Wang H, Lin X, Xu M, Lan W, Wang J. Harnessing natural saponins: Advancements in mitochondrial dysfunction and therapeutic applications. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156383. [PMID: 39848019 DOI: 10.1016/j.phymed.2025.156383] [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: 11/06/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Mitochondrial dysfunction plays a crucial role in the development of a variety of diseases, notably neurodegenerative disorders, cardiovascular diseases, metabolic syndrome, and cancer. Natural saponins, which are intricate glycosides characterized by steroidal or triterpenoid structures, have attracted interest due to their diverse pharmacological benefits, including anti-inflammatory, antiviral, and anti-aging effects. PURPOSE This review synthesizes recent advancements in understanding mitochondrial dysfunction and explores how saponins can modulate mitochondrial function. It focuses on their potential applications in neuroprotection, cardiovascular health, and oncology. STUDY DESIGN The review incorporates a comprehensive literature analysis, highlighting the interplay between saponins and mitochondrial signaling pathways. Specific attention is given to the effects of saponins like ginsenoside Rg2 and 20(S)-protopanaxatriol on mitophagy and their neuroprotective, anti-aging, and synergistic therapeutic effects when combined. METHODS We conducted a comprehensive review of current research and clinical trials using PubMed, Google Scholar, and SciFinder databases. The search focused on saponins' role in mitochondrial function and their therapeutic effects, including "saponins", "mitochondria" and "mitochondrial function". The analysis primarily focused on articles published between 2011 and 2024. RESULTS The findings indicate that certain saponins can enhance mitophagy and modulate mitochondrial signaling pathways, showing promise in neuroprotection and anti-aging. Additionally, combinations of saponins have demonstrated synergistic effects in myocardial protection and cancer therapy, potentially improving therapeutic outcomes. CONCLUSION Although saponins exhibit significant potential in modulating mitochondrial functions and developing innovative therapeutic strategies, their clinical applications are constrained by low bioavailability. Rigorous clinical trials are essential to translate these findings into effective clinical therapies, ultimately improving patient outcomes through a deeper understanding of saponins' impact on mitochondrial function.
Collapse
Affiliation(s)
- Hongmei Liu
- Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Department of pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610000, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinyu Lin
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Min Xu
- Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Department of pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610000, China
| | - Wenying Lan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinlian Wang
- Traditional Chinese Medicine Hospital of Meishan, Meishan 620010, China.
| |
Collapse
|
23
|
Wang Y, Ge Y, Hua S, Shen C, Cai B, Zhao H. Aloe-Emodin Improves Mitophagy in Alzheimer's Disease via Activating the AMPK/PGC-1α/SIRT3 Signaling Pathway. CNS Neurosci Ther 2025; 31:e70346. [PMID: 40125832 PMCID: PMC11931456 DOI: 10.1111/cns.70346] [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: 12/19/2024] [Revised: 02/22/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Impaired mitophagy results in the accumulation of defective mitochondria that are unable to be cleared effectively in Alzheimer's disease (AD). Aloe-emodin (AE), a key component of the traditional Chinese medicine Rhubarb, exhibits neuroprotective effects against Alzheimer's disease, though the underlying mechanism remains unclear. Studying aloe-emodin's role in enhancing mitophagy is vital for improving cognitive function and reducing neuronal damage in Alzheimer's disease. METHODS The APP/PS1 double transgenic mice were adopted as models for AD to assess the effects of aloe-emodin upon cognitive function and its neuroprotective impact on hippocampal neurons. Additionally, we investigated the regulatory mechanisms of proteins within the aforementioned pathway, and the morphological characteristics of mitophagy-related proteins. An AD hippocampal neuron model was developed using Aβ25-35 to evaluate the mitochondrial function, the protein expression of such a pathway and the mitophagy. This approach aims to elucidate the effects and underlying mechanisms of aloe-emodin in relation to AD. RESULTS AE activates mitophagy in neurons, improves cognitive dysfunction, reduces hippocampal damage, and alleviates AD symptoms in model mice. AE activates the expression of AMPK, PGC-1α and SIRT3. Increased expression of SIRT3 in mitochondria promotes mitophagy and regulates the function of mitochondrial proteins. When mitochondrial autophagy is enhanced, the expression of Beclin1, LC3, P62, Parkin, and PINK1-related proteins changes. Further in vitro experiments showed that AE can enhance mitochondrial function in Alzheimer's disease cell models. The mitochondrial membrane potential, GSH, ROS and Ca2+ levels gradually recover, alleviating the pathological manifestations of AD. Knocking down SIRT3 leads to increased mitochondrial damage and a reduction in mitophagy in HT22 cells. CONCLUSION Experimental results show that AE can activate mitophagy through AMPK/PGC-1α/SIRT3 pathway, alleviate cognitive dysfunction in AD, and reduce damage to hippocampal neurons.
Collapse
Affiliation(s)
- Yulu Wang
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Yunzhi Ge
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Siyu Hua
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Chenrui Shen
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
| | - Biao Cai
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
- Institute of Integrated Chinese and Western MedicineAnhui Academy of Chinese MedicineHefeiChina
- Key Laboratory of Xin'an MedicineAnhui University of Chinese Medicine, Ministry of EducationHefeiChina
- Anhui Province Key Laboratory of Chinese Medicinal FormulaHefeiChina
| | - Han Zhao
- College of Integrated Chinese and Western MedicineAnhui University of Chinese MedicineHefeiChina
- Institute of Integrated Chinese and Western MedicineAnhui Academy of Chinese MedicineHefeiChina
- Key Laboratory of Xin'an MedicineAnhui University of Chinese Medicine, Ministry of EducationHefeiChina
- Anhui Province Key Laboratory of Chinese Medicinal FormulaHefeiChina
| |
Collapse
|
24
|
Reho P, Kalia V, Jackson GL, Wang F, Eiten E, Brennan K, Brickman AM, Mayeux R, Miller GW, Vardarajan BN, Baccarelli A, Wu H. Preclinical Alzheimer's disease shows alterations in circulating neuronal-derived extracellular vesicle microRNAs in a multiethnic cohort. Alzheimers Dement 2025; 21:e70050. [PMID: 40042514 PMCID: PMC11881609 DOI: 10.1002/alz.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/21/2025] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the leading cause of dementia, affecting around 50 million individuals worldwide. Brain-derived extracellular vesicles (EVs) can cross the blood-brain barrier carrying neuron-specific molecules, such as microRNAs (miRNAs), which have potential as biomarkers of neurodegeneration. METHODS We explored the association between neuronal-derived EV miRNAs from serum and AD clinical status by performing a transcriptome-wide association study involving 46 participants with clinical AD, 14 participants with preclinical AD, and 60 neurologically healthy controls. RESULTS We identified 14 miRNAs associated with AD risk, with more pronounced transcriptional alterations in preclinical individuals compared to clinical AD individuals. Functional analysis revealed enrichment of miRNA-target genes in neurodegenerative pathways, highlighting synuclein alpha (SNCA), cytochrome c, somatic (CYCS), and microtubule associated protein tau (MAPT) as key targets. DISCUSSION Our results highlight the potential role of neuronal-derived EVs in neurodegeneration and suggest avenues for further research into brain-derived biomarkers. HIGHLIGHTS Neuronal-derived extracellular vesicles (NDEVs) carry potential brain biomarkers. We tested the association between NDEV microRNAs (miRNAs) and Alzheimer's disease (AD). Fourteen NDEV miRNAs were associated with AD. Preclinical AD displayed more pronounced transcriptional changes than clinical AD. miRNA-target genes were enriched in pathways associated with neurodegeneration.
Collapse
Affiliation(s)
- Paolo Reho
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Gabriela L. Jackson
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Fang Wang
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | | | - Kasey Brennan
- Department of Environmental Health, Harvard T. H. Chan School of Public HealthHarvard UniversityBostonMassachusettsUSA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude. H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, College of Physicians and SurgeonsColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude. H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, College of Physicians and SurgeonsColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of Epidemiology, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Badri N. Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude. H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, College of Physicians and SurgeonsColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Andrea Baccarelli
- Department of Environmental Health, Harvard T. H. Chan School of Public HealthHarvard UniversityBostonMassachusettsUSA
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
25
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
26
|
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
|
27
|
de Lima EP, Laurindo LF, Catharin VCS, Direito R, Tanaka M, Jasmin Santos German I, Lamas CB, Guiguer EL, Araújo AC, Fiorini AMR, Barbalho SM. Polyphenols, Alkaloids, and Terpenoids Against Neurodegeneration: Evaluating the Neuroprotective Effects of Phytocompounds Through a Comprehensive Review of the Current Evidence. Metabolites 2025; 15:124. [PMID: 39997749 PMCID: PMC11857241 DOI: 10.3390/metabo15020124] [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: 12/24/2024] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/26/2025] Open
Abstract
Neurodegenerative diseases comprise a group of chronic, usually age-related, disorders characterized by progressive neuronal loss, deformation of neuronal structure, or loss of neuronal function, leading to a substantially reduced quality of life. They remain a significant focus of scientific and clinical interest due to their increasing medical and social importance. Most neurodegenerative diseases present intracellular protein aggregation or their extracellular deposition (plaques), such as α-synuclein in Parkinson's disease and amyloid beta (Aβ)/tau aggregates in Alzheimer's. Conventional treatments for neurodegenerative conditions incur high costs and are related to the development of several adverse effects. In addition, many patients are irresponsive to them. For these reasons, there is a growing tendency to find new therapeutic approaches to help patients. This review intends to investigate some phytocompounds' effects on neurodegenerative diseases. These conditions are generally related to increased oxidative stress and inflammation, so phytocompounds can help prevent or treat neurodegenerative diseases. To achieve our aim to provide a critical assessment of the current literature about phytochemicals targeting neurodegeneration, we reviewed reputable databases, including PubMed, EMBASE, and COCHRANE, seeking clinical trials that utilized phytochemicals against neurodegenerative conditions. A few clinical trials investigated the effects of phytocompounds in humans, and after screening, 13 clinical trials were ultimately included following PRISMA guidelines. These compounds include polyphenols (flavonoids such as luteolin and quercetin, phenolic acids such as rosmarinic acid, ferulic acid, and caffeic acid, and other polyphenols like resveratrol), alkaloids (such as berberine, huperzine A, and caffeine), and terpenoids (such as ginkgolides and limonene). The gathered evidence underscores that quercetin, caffeine, ginkgolides, and other phytochemicals are primarily anti-inflammatory, antioxidant, and neuroprotective, counteracting neuroinflammation, neuronal oxidation, and synaptic dysfunctions, which are crucial aspects of neurodegenerative disease intervention in various included conditions, such as Alzheimer's and other dementias, depression, and neuropsychiatric disorders. In summary, they show that the use of these compounds is related to significant improvements in cognition, memory, disinhibition, irritability/lability, aberrant behavior, hallucinations, and mood disorders.
Collapse
Affiliation(s)
- Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Vitor Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos Krt. 113, H-6725 Szeged, Hungary
| | - Iris Jasmin Santos German
- Department of Biological Sciences (Anatomy), School of Dentistry of Bauru, University of São Paulo (FOB-USP), Alameda Doutor Octávio Pinheiro Brisolla, 9-75, Bauru 17012-901, São Paulo, Brazil
| | - Caroline Barbalho Lamas
- Department of Gerontology, School of Gerontology, Universidade Federal de São Carlos (UFSCar), São Carlos 13565-905, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Adriana Maria Ragassi Fiorini
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| |
Collapse
|
28
|
Chiang MC, Yang YP, Nicol CJB, Chiang T, Yen C. Resveratrol-Enhanced Human Neural Stem Cell-Derived Exosomes Mitigate MPP+-Induced Neurotoxicity Through Activation of AMPK and Nrf2 Pathways and Inhibition of the NLRP3 Inflammasome in SH-SY5Y Cells. Life (Basel) 2025; 15:294. [PMID: 40003703 PMCID: PMC11856727 DOI: 10.3390/life15020294] [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: 12/26/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder primarily characterized by the loss of dopaminergic neurons in the substantia nigra. Mitochondrial dysfunction, oxidative stress, and neuroinflammation are recognized as critical pathological mechanisms driving neurodegeneration in PD. Exosome (Exo)-based therapies, particularly those derived from human neural stem cells (hNSCs), offer promising neuroprotective effects due to their ability to transfer bioactive molecules that modulate cellular processes. Resveratrol (RES), a polyphenolic compound with potent antioxidant and anti-inflammatory properties, has been shown to enhance the therapeutic potential of stem cell (SC)-derived Exos. This study investigated the neuroprotective effects of RES-treated hNSCs-derived Exos (RES-hNSCs-Exos) on SH-SY5Y cells exposed to 1-methyl-4-phenylpyridinium (MPP+), a neurotoxin commonly used to model Parkinsonian neurotoxicity. Treating SH-SY5Y cells with MPP+ led to significant reductions in cell viability, mitochondrial dysfunction, increased oxidative stress, and the activation of inflammatory pathways. Treatment with RES-hNSCs-Exos rescued SH-SY5Y cells from MPP+-induced toxicity by improving cell viability, enhancing ATP production, increasing mitochondrial biogenesis, and reducing reactive oxygen species (ROS) generation. The findings also demonstrated the increased expression of essential genes involved in mitochondrial biogenesis, such as PGC1α, NRF1, and Tfam, indicating improved mitochondrial function in the presence of RES-hNSCs-Exos. Further analysis revealed that these protective effects were mediated by activating the AMP-activated protein kinase (AMPK) and Nrf2 signaling pathways, which promoted mitochondrial health and reduced oxidative stress. Moreover, RES-hNSCs-Exos treatment suppressed neuroinflammation by downregulating NLRP3 inflammasome activation and reducing the secretion of pro-inflammatory cytokines IL-1β and IL-18. In conclusion, the results suggest that RES-hNSCs-Exos exhibit potent neuroprotective effects against MPP+-induced neurotoxicity by enhancing mitochondrial function, reducing oxidative stress, and inhibiting neuroinflammation. These findings highlight the potential of hNSCs-Exos as a novel therapeutic strategy for neurodegenerative diseases like PD, with RES as a valuable enhancer of Exos efficacy.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Christopher J. B. Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, and Cancer Biology and Genetics Division, Sinclair Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Tairui Chiang
- Ames Middle School, Ames, IA 50014, USA
- New Taipei Municipal Jinhe High School, New Taipei City 235, Taiwan
| | - Chiahui Yen
- Department of International Business, Ming Chuan University, Taipei 111, Taiwan
| |
Collapse
|
29
|
Meng T, Zhang Y, Ye Y, Li H, He Y. Bioinformatics insights into mitochondrial and immune gene regulation in Alzheimer's disease. Eur J Med Res 2025; 30:89. [PMID: 39920860 PMCID: PMC11806906 DOI: 10.1186/s40001-025-02297-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: 03/27/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND There is growing evidence that the pathogenesis of Alzheimer's disease is closely linked to the resident innate immune cells of the central nervous system, including microglia and astrocytes. Mitochondrial dysfunction in microglia has also been reported to play an essential role in the pathogenesis of AD and other neurological diseases. Therefore, finding the mitochondrial and immune-related gene (MIRG) signatures in AD can be significant in diagnosing and treating AD. METHODS In this study, the intersection of the differentially expressed genes (DEGs) from the GSE109887 cohort, immune-related genes (IRGs) obtained from WGCNA analysis, and mitochondria-related genes (MRGs) was taken to identify mitochondria-immune-related genes (MIRGs). Then, using machine learning algorithms, biomarkers with good diagnostic value were selected, and a nomogram was constructed. Subsequently, we further analyzed the signaling pathways and potential biological mechanisms of the biomarkers through gene set enrichment analysis, prediction of transcription factors (TFs), miRNAs, and drug prediction. RESULTS Using machine learning algorithms, five biomarkers (TSPO, HIGD1A, NDUFAB1, NT5DC3, and MRPS30) were successfully identified, and a nomogram model with strong diagnostic ability and accuracy (AUC > 0.9) was constructed. In addition, single-gene enrichment analysis revealed that NDUFAB1 was significantly enriched in pathways associated with diseases, such as Alzheimer's and Parkinson's, providing valuable insights for future clinical research on Alzheimer's in the context of mitochondrial-immune interactions. Interestingly, brain tissue pathology showed neuronal atrophy and demyelination in AD mice, along with a reduction in Nissl bodies. Furthermore, the escape latency of AD mice was significantly longer than that of the control group. After platform removal, there was a notable increase in the path complexity and time required to reach the target quadrant, suggesting a reduction in spatial memory capacity in AD mice. Moreover, qRT-PCR validation confirmed that the mRNA expression of the five biomarkers was consistent with bioinformatics results. In AD mice, TSPO expression was increased, while HIGD1A, NDUFAB1, NT5DC3, and MRPS30 expressions were decreased. However, peripheral blood samples did not show expression of HIGD1A or MRPS30. These findings provide new insights for research on Alzheimer's disease in the context of mitochondrial-immune interactions, further exploring the pathogenesis of Alzheimer's disease and offering new perspectives for the clinical development of novel drugs. CONCLUSIONS Five mitochondrial and immune biomarkers, i.e., TSPO, HIGD1A, NDUFAB1, NT5DC3, and MRPS30, with diagnostic value in Alzheimer's disease, were screened by machine-learning algorithmic models, which will be a guide for future clinical research of Alzheimer's disease in the mitochondria-immunity-related direction.
Collapse
Affiliation(s)
- Tian Meng
- Yunnan Yunke Institute of Biotechnology, No. 871 Longquan Rd, Kunming, 650500, China
| | - Yazhou Zhang
- Department of Geriatrics, The Second People's Hospital of Kunming, No. 338Guangming Rd, Kunming, 650233, Yunnan, China
| | - Yuan Ye
- Department of Geriatrics, The Second People's Hospital of Kunming, No. 338Guangming Rd, Kunming, 650233, Yunnan, China
| | - Hui Li
- Yunnan Labreal Biotechnology Co., LTD, No. 871 Longquan Rd, Kunming, 650500, China
| | - Yongsheng He
- Yunnan Yunke Institute of Biotechnology, No. 871 Longquan Rd, Kunming, 650500, China.
- Yunnan Labreal Biotechnology Co., LTD, No. 871 Longquan Rd, Kunming, 650500, China.
| |
Collapse
|
30
|
Spina E, Ferrari RR, Pellegrini E, Colombo M, Poloni TE, Guaita A, Davin A. Mitochondrial Alterations, Oxidative Stress, and Therapeutic Implications in Alzheimer's Disease: A Narrative Review. Cells 2025; 14:229. [PMID: 39937020 PMCID: PMC11817193 DOI: 10.3390/cells14030229] [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: 12/17/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
The relationship between aging, mitochondrial dysfunction, neurodegeneration, and the onset of Alzheimer's disease (AD) is a complex area of study. Aging is the primary risk factor for AD, and it is associated with a decline in mitochondrial function. This mitochondrial dysfunction is believed to contribute to the neurodegenerative processes observed in AD. Neurodegeneration in AD is characterized by the progressive loss of synapses and neurons, particularly in regions of the brain involved in memory and cognition. It is hypothesized that mitochondrial dysfunction plays a pivotal role by disrupting cellular energy metabolism and increasing the production of reactive oxygen species (ROS), which can damage cellular components and exacerbate neuronal loss. Despite extensive research, the precise molecular pathways linking mitochondrial dysfunction to AD pathology are not fully understood. Various hypotheses have been proposed, including the mitochondrial cascade hypothesis, which suggests that mitochondrial dysfunction is an early event in AD pathogenesis that triggers a cascade of cellular events leading to neurodegeneration. With this narrative review, we aim to summarize some specific issues in the literature on mitochondria and their involvement in AD onset, with a focus on the development of therapeutical strategies targeting the mitochondria environment and their potential application for the treatment of AD itself.
Collapse
Affiliation(s)
- Erica Spina
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (E.S.); (E.P.); (M.C.); (A.G.); (A.D.)
| | - Riccardo Rocco Ferrari
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (E.S.); (E.P.); (M.C.); (A.G.); (A.D.)
- Department of Brain and Behavioral Sciences, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy
| | - Elisa Pellegrini
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (E.S.); (E.P.); (M.C.); (A.G.); (A.D.)
| | - Mauro Colombo
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (E.S.); (E.P.); (M.C.); (A.G.); (A.D.)
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy;
| | - Antonio Guaita
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (E.S.); (E.P.); (M.C.); (A.G.); (A.D.)
| | - Annalisa Davin
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (E.S.); (E.P.); (M.C.); (A.G.); (A.D.)
| |
Collapse
|
31
|
Joshi DC, Chavan MB, Gurow K, Gupta M, Dhaliwal JS, Ming LC. The role of mitochondrial dysfunction in Huntington's disease: Implications for therapeutic targeting. Biomed Pharmacother 2025; 183:117827. [PMID: 39854819 DOI: 10.1016/j.biopha.2025.117827] [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: 10/11/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Huntington's disease (HD) is a progressive, autosomal dominant neurodegenerative disorder characterized by cognitive decline, motor dysfunction, and psychiatric disturbances. A common feature of neurodegenerative disorders is mitochondrial dysfunction, which affects the brain's sensitivity to oxidative damage and its high oxygen demand. This dysfunction may plays a significant role in the pathogenesis of Huntington's disease. HD is caused by a CAG repeat expansion in the huntingtin gene, which leads to the production of a toxic mutant huntingtin (mHTT) protein. This disruption in mitochondrial function compromises energy metabolism and increases oxidative stress, resulting in mitochondrial DNA abnormalities, impaired calcium homeostasis, and altered mitochondrial dynamics. These effects ultimately may contribute to neuronal dysfunction and cell death, underscoring the importance of targeting mitochondrial function in developing therapeutic strategies for HD. This review discusses the mechanistic role of mitochondrial dysfunction in Huntington's disease. Mitochondrial dysfunction is a crucial factor in HD, making mitochondrial-targeted therapies a promising approach for treatment. We explore therapies that address bioenergy deficits, antioxidants that reduce reactive oxygen species, calcium modulators that restore calcium homeostasis, and treatments that enhance mitochondrial dynamics to rejuvenate mitochondrial function. We also highlight innovative treatment approaches such as gene editing and stem cell therapy, which offer hope for more personalized strategies. In conclusion, understanding mitochondrial dysfunction in Huntington's disease may guide potential treatment strategies. Targeting this dysfunction may help to slow disease progression and enhance the quality of life for individuals affected by Huntington's disease.
Collapse
Affiliation(s)
- Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Dist., Ajmer, Rajasthan, India.
| | - Mayuri Bapu Chavan
- TMV's Lokmanya Tilak Institute of Pharmaceutical Sciences, Pune, Maharashtra, India.
| | - Kajal Gurow
- Department of Pharmacology, Gurukul Pharmacy college, Ranpur, Kota, Rajasthan, India
| | - Madhu Gupta
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India.
| | | | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia; Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (deemed to be University), Sawangi (M), Wardha, India.
| |
Collapse
|
32
|
Muraleva NA, Zhdankina AA, Khlebnikov AI, Kolosova NG. JNK Signaling Pathway Activity Alterations in the Rat Hippocampus: Effect of Age, Alzheimer's Disease-Like Pathology Development, and the JNK Inhibitor IQ-1S. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:265-275. [PMID: 40254404 DOI: 10.1134/s0006297924603903] [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: 10/28/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 04/22/2025]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder and the leading cause of senile dementia. The key risk factor for a more common (>95% of cases) sporadic form of AD is age. So far, there are no effective methods for AD prevention or treatment. A growing body of evidence indicates that the development of AD and other neurodegenerative diseases is associated with the activation of mitogen-activated protein kinase (MAPK) pathways, and JNK signaling pathway is considered as a potential target for the prevention and treatment of AD. However, the information on alterations in its activity in ontogenesis, which are evaluated by changes in the phosphorylation of its components, is extremely limited. The aim of this study was to compare age-related changes in the activity of JNK signaling pathway in the hippocampus of Wistar rats and senescence-accelerated OXYS rats (which spontaneously develop the key symptoms of AD-like pathology) and to evaluate the effect of the selective JNK3 inhibitor IQ-1S (11H-indeno[1,2-b]quinoxalin-11-one oxime sodium salt). The ability of IQ-1S to suppress accelerated brain aging in OXYS rat has been proven previously, but the effect of this inhibitor on the JNK activity has not been studied. Here, we showed that with age, the activity of the JNK signaling pathway increased in the hippocampus of rats of both strains. At the same time, the manifestation and active progression of AD-like pathology in OXYS rats was accompanied by the increase in the phosphorylation level of the key kinase of this signaling pathway, JNK3, and its target proteins compared to Wistar rats, which allowed us to suggest JNK3 as a potential target for interventions aimed at preventing neurodegenerative processes. This suggestion was supported by the fact that the neuroprotective effect of the selective JNK3 inhibitor IQ-1S and its ability to suppress the development of neurodegenerative processes in OXYS rats were associated with a decrease in the phosphorylation levels of JNK3, c-Jun, APP, and Tau in the hippocampus.
Collapse
Affiliation(s)
- Natalia A Muraleva
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | | | - Andrey I Khlebnikov
- Kizhner Research Center, Tomsk Polytechnic University, Tomsk, 634050, Russia
| | - Nataliya G Kolosova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| |
Collapse
|
33
|
Cui H, Li Z, Sun H, Zhao W, Ma H, Hao L, Zhang Z, Hölscher C, Ma D, Zhang Z. The neuroprotective effects of cholecystokinin in the brain: antioxidant, anti-inflammatory, cognition, and synaptic plasticity. Rev Neurosci 2025:revneuro-2024-0142. [PMID: 39832348 DOI: 10.1515/revneuro-2024-0142] [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: 10/07/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
Cholecystokinin (CCK) is a major neuropeptide in the brain that functions as a neurotransmitter, hormone, and growth factor. The peptide and its receptors are widely expressed in the brain. CCK signaling modulates synaptic plasticity and can improve or impair memory formation, depending on the brain areas studies and the receptor subtype activated. Studies have shown in a series of animal models of neurodegenerative diseases that CCK receptor agonists show neuroprotective effects and can effectively alleviate oxidative stress, alleviate chronic inflammation of the central nervous system, improve neuronal synaptic plasticity, prevent neuronal loss, and improve cognitive dysfunction in Alzheimer's disease (AD) model mice and motor activity in animal models of Parkinson's disease. In addition, CCK plays important roles in the amygdala to regulate anxiety and depressive states. Activation of interneurons or inhibition of excitatory neurons can improve anxiety levels. This review summarizes the effects on memory formation and synaptic plasticity, the neuroprotective effects of cholecystokinin and its analogs in neurological diseases such as Alzheimer and Parkinson's disease, and the effects on anxiety and neuronal activity in the amygdala.
Collapse
Affiliation(s)
- Hailiang Cui
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Zhonghua Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hongyu Sun
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Wanlin Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - He Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Li Hao
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Christian Hölscher
- Henan Academy of Innovations in Medical Science, Brain Institute, Zhengzhou 451100, Henan Province, China
| | - Dongrui Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Zijuan Zhang
- School of Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| |
Collapse
|
34
|
Wang T, Huang X, Sun S, Wang Y, Han L, Zhang T, Zhang T, Chen X. Recent Advances in the Mechanisms of Postoperative Neurocognitive Dysfunction: A Narrative Review. Biomedicines 2025; 13:115. [PMID: 39857699 PMCID: PMC11762480 DOI: 10.3390/biomedicines13010115] [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: 12/03/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Postoperative neurocognitive dysfunction (PND) is a prevalent and debilitating complication in elderly surgical patients, characterized by persistent cognitive decline that negatively affects recovery and quality of life. As the aging population grows, the rising number of elderly surgical patients has made PND an urgent clinical challenge. Despite increasing research efforts, the pathophysiological mechanisms underlying PND remain inadequately characterized, underscoring the need for a more integrated framework to guide targeted interventions. To better understand the molecular mechanisms and therapeutic targets of PND, this narrative review synthesized evidence from peer-reviewed studies, identified through comprehensive searches of PubMed, Embase, Cochrane Library, and Web of Science. Key findings highlight neuroinflammation, oxidative stress, mitochondrial dysfunction, neurotransmitter imbalances, microvascular changes, and white matter lesions as central to PND pathophysiology, with particular parallels to encephalocele- and sepsis-associated cognitive impairments. Among these, neuroinflammation, mediated by pathways such as the NLRP3 inflammasome and blood-brain barrier disruption, emerges as a pivotal driver, triggering cascades that exacerbate neuronal injury. Oxidative stress and mitochondrial dysfunction synergistically amplify these effects, while neurotransmitter imbalances and microvascular alterations, including white matter lesions, contribute to synaptic dysfunction and cognitive decline. Anesthetic agents modulate these interconnected pathways, exhibiting both protective and detrimental effects. Propofol and dexmedetomidine demonstrate neuroprotective properties by suppressing neuroinflammation and microglial activation, whereas inhalational anesthetics like sevoflurane intensify oxidative stress and inflammatory responses. Ketamine, with its anti-inflammatory potential, offers promise but requires further evaluation to determine its long-term safety and efficacy. By bridging molecular insights with clinical practice, this review highlights the critical role of personalized anesthetic strategies in mitigating PND and improving cognitive recovery in elderly surgical patients. It aims to inform future research and clinical decision-making to address this multifaceted challenge.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xin Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (T.W.); (X.H.); (S.S.); (Y.W.); (L.H.); (T.Z.); (T.Z.)
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of Education, Wuhan 430022, China
| |
Collapse
|
35
|
Ergüç A, Albayrak G, Muhammed MT, Karakuş F, Arzuk E. Mitochondrial toxicity of selected natural compounds: in vitro assessment and in silico molecular docking and dynamics simulation. Drug Chem Toxicol 2025; 48:199-209. [PMID: 39411844 DOI: 10.1080/01480545.2024.2412775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 02/07/2025]
Abstract
Prangos uechtritzii Boiss & Hausskn stands out for its rich bioactive constituents including prantschimgin (PRA), imperatorin (IMP), suberosin (SUB), adicardin (ADI), and oxypeucedanin hydrate (OPH) in the Apiaceae family. Although these molecules contribute to several biological activities, their mitochondrial toxicity were not illuminated in depth with the appropriate in vitro and in silico models. Cell viability studies investigated the cytotoxic activities of molecules in HepG2 cells by replacing glucose with galactose due to Warburg effects. Mitochondrial toxicity (mitotoxicity) parameters such as cellular adenosine triphosphate (ATP) and mitochondrial membrane potential (MMP) levels were assessed with cytotoxic concentrations of selected molecules. Molecular docking and dynamics studies were also conducted against mitochondrial electron transport chain (ETC) complexes (I-V) with selected compounds. In vitro results showed that PRA, SUB, and IMP reduced cell viability more in galactose media compared to high glucose media in a dose-dependent manner. PRA, IMP, and SUB decreased ATP levels and MMP, especially in the galactose medium. The in silico study revealed that PRA, IMP, and SUB might bind to complexes I-V at different levels. The docking study demonstrated that PRA had the highest binding potential with the complexes, higher than the standard ligands in some cases. The molecular dynamics (MD) simulation study showed that PRA formed stable complexes with complexes II, III, and IV. In addition, PRA was anticipated to remain inside the binding site of complex II most stably during the 230 ns simulation period. Our study suggests that PRA, IMP, and SUB exhibit mitotoxicity.
Collapse
Affiliation(s)
- Ali Ergüç
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ondokuz Mayıs University, Samsun, Türkiye
| | - Gökay Albayrak
- Department of Pharmaceutical Botany, Faculty of Pharmacy, İzmir Katip Çelebi University, İzmir, Türkiye
| | - Muhammed Tilahun Muhammed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Süleyman Demirel University, Isparta, Türkiye
| | - Fuat Karakuş
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Van Yuzuncu Yil University, Van, Türkiye
| | - Ege Arzuk
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ege University, İzmir, Türkiye
| |
Collapse
|
36
|
Deng L, Luo Q, Liu Y, Wang Y, Xiong Z, Wang H, Zhao L, Jia L, Shi R, Huang C, Chen Z. Progressive iron overload in middle-aged mice impairs olfactory function, triggers lipid oxidation and induces apoptosis. Front Pharmacol 2024; 15:1506944. [PMID: 39749201 PMCID: PMC11693683 DOI: 10.3389/fphar.2024.1506944] [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: 10/06/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction This study aims to investigate the progressive impact of chronic iron overload on the olfactory bulb, a region significantly affected in early neurodegenerative diseases like Parkinson's and Alzheimer's. The focus is on understanding how iron accumulation leads to oxidative stress, mitochondrial dysfunction, and neuronal damage over time in middle-aged mice. Method The mice were continuously administered FC for a duration of 16 weeks, and the olfactory behavior of the mice was observed at intervals of 4 weeks. Inductively coupled plasma mass spectrometry (ICP-MS) was employed to detect alterations in iron content within the olfactory bulb of the mice, while levels of lipid peroxidation and antioxidant indexes were assessed using biochemical kits. Additionally, western blotting and qPCR techniques were utilized to analyze transcriptional and expression changes in proteins and genes related to iron metabolism. Furthermore, microstructural modifications as well as mitochondrial observations were conducted through paraffin sectioning and transmission electron microscopy (TEM). Result A significant and progressive increase in iron accumulation in the olfactory bulb, starting from week 8 and peaking at week 16. This accumulation coincided with a decline in olfactory function observed at week 12. Key markers of oxidative stress, such as 4-HNE and MDA, were elevated in specific layers, and antioxidant defenses were reduced. Mitochondrial damage became evident from week 8, with caspase-3 activation indicating increased apoptosis, particularly in the granular layer. This study is to demonstrate the link between chronic iron overload and progressive olfactory dysfunction in the context of neurodegenerative diseases. It provides evidence that iron-induced oxidative stress and mitochondrial damage in the olfactory bulb contribute to early sensory deficits, suggesting that the olfactory bulb's selective vulnerability can serve as an early biomarker for neurodegenerative conditions. Conclusion Chronic iron overload leads to progressive oxidative damage, mitochondrial dysfunction, and apoptosis in the olfactory bulb, causing sensory deficits. Targeting iron accumulation and oxidative damage may offer new strategies for early intervention in neurodegenerative diseases, highlighting the importance of addressing iron dysregulation.
Collapse
Affiliation(s)
- Lin Deng
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
- Safety Evaluation Center, Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), Chengdu, China
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Yucong Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Yao Wang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Zongliang Xiong
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Hongping Wang
- Safety Evaluation Center, Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), Chengdu, China
| | - Lu Zhao
- Safety Evaluation Center, Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), Chengdu, China
| | - Lanlan Jia
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Riyi Shi
- Department of Basic Medical Sciences, Center for Paralysis Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, China
| |
Collapse
|
37
|
Keighron CN, Avazzedeh S, Quinlan LR. Robust In Vitro Models for Studying Parkinson's Disease? LUHMES Cells and SH-SH5Y Cells. Int J Mol Sci 2024; 25:13122. [PMID: 39684831 DOI: 10.3390/ijms252313122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
As our population ages, there is an increased unmet clinical need surrounding neurodegenerative diseases such as Parkinson's disease (PD). To tackle this ever-increasing problem, we must ensure that the cell models that we use to develop therapeutics in vitro are robust, reliable, and replicable. In this study, we compared SH-SY5Y cells with LUHMES cells in response to 6-Hydroxydopamine (6OHDA) and 1-Methyl-4-phenylpyridinium (MPP+), two common Parkinson's insults used in in vitro analysis. Both these cell types have apparent dopaminergic phenotypes, which could aid us in understanding their potential in this race to novel therapies. The LUHMES cells showed consistent dopaminergic (DA) expression through tyrosine hydroxylase (TH) positivity, alongside depleted ATP levels and elevated reactive oxygen species (ROS) production, whereas the SH-SH5Y cells displayed resilience to both chemical insults, raising questions about their utility in accurately modelling PD pathology. Further electrophysiological analysis revealed comparable firing rates and ion channel signalling between both cell types; however, LUHMES cells demonstrated stronger calcium signalling responses, further supporting their use as a more robust PD model. While SH-SY5Y cells showed some adaptability in vitro, their inconsistent DA phenotype and limited response to chemical insults limit their suitability for advanced research, suggesting that LUHMES cells should and must take their place as a hallmark in Parkinson's disease research.
Collapse
Affiliation(s)
- Cameron Noah Keighron
- Cellular Physiology Research Lab, School of Medicine, Department of Physiology, University of Galway, H91W5P7 Galway, Ireland
| | - Sahar Avazzedeh
- Cellular Physiology Research Lab, School of Medicine, Department of Physiology, University of Galway, H91W5P7 Galway, Ireland
| | - Leo R Quinlan
- Cellular Physiology Research Lab, School of Medicine, Department of Physiology, University of Galway, H91W5P7 Galway, Ireland
- CÚRAM SFI Research Centre for Medical Devices, University of Galway, H91W2TY Galway, Ireland
| |
Collapse
|
38
|
Shang Y, Chen K, Ni H, Zhu X, Yuan X, Wang Y, Liu X, Cui Z, Niu Y, Shi Y, Wu H, Xia D, Wu Y. Environmentally relevant concentrations of perfluorobutane sulfonate impair locomotion behaviors and healthspan by downregulating mitophagy in C. elegans. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:135938. [PMID: 39326150 DOI: 10.1016/j.jhazmat.2024.135938] [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: 07/27/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Perfluorobutane sulfonate (PFBS), a chemical compound within the group of per- and polyfluoroalkyl substances (PFAS), has been utilized as an alternative to perfluorooctane sulfonate (PFOS) recently. Previous research has indicated that PFBS might be linked to a range of health concerns. However, the potential impacts of environmentally relevant concentrations of PFBS (25 nM) on aging as well as the underlying mechanisms remained largely unexplored. In this study, we investigated the impact of PFBS exposure on aging and the associated mechanisms in Caenorhabditis elegans. Our findings indicated that exposure to PFBS impaired healthspan of C. elegans. Through bioinformatic screening analyses, we identified that the dysfunctions of pink-1 mediated mitophagy might play a critical role in PFBS induced aging. The results furtherly revealed that PFBS exposure led to elevated levels of reactive oxygen species (ROS) and mitophagy impairment through downregulating pink-1/pdr-1 pathway. Furthermore, the mitophagy agonist Urolithin A (UA) effectively reversed PFBS-induced mitophagy dysfunction and enhanced healthspan in C. elegans. Taken together, our study suggested that exposure to environmentally relevant concentrations of PFBS could accelerate aging by downregulating the pink-1 mediated mitophagy. Promoting mitophagy within cells could be a promising therapeutic strategy for delaying PFBS-induced aging.
Collapse
Affiliation(s)
- Yahui Shang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelie Chen
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gynecology and Obstetrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Ni
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Zhu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Yuan
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuwei Wang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinxin Liu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenyan Cui
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuequn Niu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Yu Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Han Wu
- Department of Ophthalmology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dajing Xia
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yihua Wu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
39
|
Ye F, Wei C, Wu A. The potential mechanism of mitochondrial homeostasis in postoperative neurocognitive disorders: an in-depth review. Ann Med 2024; 56:2411012. [PMID: 39450938 PMCID: PMC11514427 DOI: 10.1080/07853890.2024.2411012] [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: 12/29/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 10/26/2024] Open
Abstract
Postoperative neurocognitive disorders (PND) are the most common neurological disorders following surgery and anaesthesia before and within 12 months after surgery, with a high prevalence in the geriatric population. PND can severely deteriorate the quality of life of patients, especially among the elderly, mainly manifested as memory loss, attention, decline and language comprehension disorders, mostly in elderly patients, with an incidence as high as 31%. Previous studies have also raised the possibility of accelerated cognitive decline and underlying neuropathological processes associated with diseases that affect cognitive performance (e.g. Alzheimer's dementia) for reasons related to anaesthesia and surgery. Currently, most research on PND has focused on various molecular pathways, especially in the geriatric population. The various hypotheses that have been proposed regarding the mechanisms imply peripheral neuroinflammation, oxidative stress, mitochondrial homeostasis, synaptic function, autophagy disorder, blood-brain barrier dysfunction, the microbiota-gut-brain axis and lack of neurotrophic support. However, the underlying pathogenesis and molecular mechanisms of PND have not yet been uncovered. Recent research has focused on mitochondrial homeostasis. In this paper, we present a review of various studies to better understand and characterize the mechanisms of associated cognitive dysfunction. As the biochemical basis of PND becomes more clearly defined, future treatments based on mitochondrial homeostasis modulation can prove to be very promising.
Collapse
Affiliation(s)
- Fan Ye
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Siddiqui S, Liu F, Kanthasamy AG, McGrail M. Stat3 mediates Fyn kinase-driven dopaminergic neurodegeneration and microglia activation. Dis Model Mech 2024; 17:dmm052011. [PMID: 39641161 PMCID: PMC11646115 DOI: 10.1242/dmm.052011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/17/2024] [Indexed: 12/07/2024] Open
Abstract
The Alzheimer's disease and Parkinson's disease risk locus FYN kinase is implicated in neurodegeneration and inflammatory signaling. To investigate in vivo mechanisms of Fyn-driven neurodegeneration, we built a zebrafish neural-specific Gal4:UAS model of constitutively active FynY531F signaling. Using in vivo live imaging, we demonstrated that neural FynY531F expression leads to dopaminergic neuron loss and mitochondrial aggregation in 5 day larval brain. Dopaminergic loss coincided with microglia activation and induction of tnfa, il1b and il12a inflammatory cytokine expression. Transcriptome analysis revealed Stat3 signaling as a potential Fyn target. Chemical inhibition experiments confirmed Fyn-driven dopaminergic neuron loss, and the inflammatory response was dependent upon activation of Stat3 and NF-κB pathways. Dual chemical inhibition demonstrated that Stat3 acts synergistically with NF-κB in dopaminergic neuron degeneration. These results identify Stat3 as a novel downstream effector of Fyn signaling in neurodegeneration and inflammation.
Collapse
Affiliation(s)
- Sahiba Siddiqui
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
- Interdepartmental Genetics and Genomics Graduate Program (IGG), Iowa State University, Ames, IA 50011, USA
| | - Fang Liu
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Anumantha G. Kanthasamy
- Center for Brain Science and Neurodegenerative Diseases, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Maura McGrail
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
- Interdepartmental Genetics and Genomics Graduate Program (IGG), Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
41
|
Yu JX, Lin M, Zhang WX, Lao FX, Huang HC. Astaxanthin Prevents Oxidative Damage and Cell Apoptosis Under Oxidative Stress Involving the Restoration of Mitochondrial Function. Cell Biochem Funct 2024; 42:e70027. [PMID: 39663588 DOI: 10.1002/cbf.70027] [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: 09/04/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Oxidative stress (OS) is one of the factors that result in cell damage and the development of neurological diseases such as Alzheimer's disease (AD). Astaxanthin (ASTA), a natural compound known for its potent antioxidant properties, shows the biological activities in anti-apoptosis and antitumor. However, its specific mechanism on mitochondrial function remains unclear. This study investigated the effects of ASTA on regulation in mitochondrial function and cell apoptosis under OS induced by hydrogen peroxide (H2O2). The results demonstrated that ASTA (0.1, 1, 10 μmol/L) protected cells form H2O2-induced cell damage and apoptosis through mitochondrial pathway. ASTA significantly reduced H2O2-induced mitochondrial dysfunctions and restored the intracellular reactive oxygen species (ROS), mitochondrial membrane potential, and respiratory capacity. These findings suggest that ASTA's antioxidant properties can benefit neurons by maintaining mitochondrial function and alleviating oxidative damage and cell apoptosis induced by H2O2.
Collapse
Affiliation(s)
- Jia-Xin Yu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Miao Lin
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Wen-Xuan Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Feng-Xue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
- Key Laboratory of Natural Products Development and Innovative Drug Research, Beijing Union University, Beijing, China
| |
Collapse
|
42
|
Gao L, Yang XN, Dong YX, Han YJ, Zhang XY, Zhou XL, Liu Y, Liu F, Fang JS, Ji JL, Gao ZR, Qin XM. The potential therapeutic strategy in combating neurodegenerative diseases: Focusing on natural products. Pharmacol Ther 2024; 264:108751. [PMID: 39522697 DOI: 10.1016/j.pharmthera.2024.108751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/25/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), Huntington disease (HD), and Multiple sclerosis (MS), pose a significant global health challenge due to their intricate pathology and limited therapeutic interventions. Natural products represent invaluable reservoirs for combating these neurodegenerative diseases by targeting key pathological hallmarks such as protein aggregation, synaptic dysfunction, aberrant proteostasis, cytoskeletal abnormalities, altered energy homeostasis, inflammation, and neuronal cell death. This review provides an in-depth analysis of the mechanisms and therapeutic targets of natural products for their neuroprotective effects. Furthermore, it elucidates the current progress of clinical trials investigating the potential of natural products in delaying neurodegeneration. The objective of this review is to enhance the comprehension of natural products in the prevention and treatment of neurodegenerative diseases, offering new insights and potential avenues for future pharmaceutical research.
Collapse
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xi-Na Yang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Yi-Xiao Dong
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Yi-Jia Han
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xin-Yue Zhang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Xin-Le Zhou
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Ying Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Fang Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China
| | - Jian-Song Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jian-Long Ji
- College of Integrated Circuits, Taiyuan University of Technology, Taiyuan, China.
| | - Zheng-Run Gao
- Songjiang Research Institute, Songjiang Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China.
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China; The Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, China.
| |
Collapse
|
43
|
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
|
44
|
Xu C, Fu X, Qin H, Yao K. Traversing the epigenetic landscape: DNA methylation from retina to brain in development and disease. Front Cell Neurosci 2024; 18:1499719. [PMID: 39678047 PMCID: PMC11637887 DOI: 10.3389/fncel.2024.1499719] [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: 09/21/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
DNA methylation plays a crucial role in development, aging, degeneration of various tissues and dedifferentiated cells. This review explores the multifaceted impact of DNA methylation on the retina and brain during development and pathological processes. First, we investigate the role of DNA methylation in retinal development, and then focus on retinal diseases, detailing the changes in DNA methylation patterns in diseases such as diabetic retinopathy (DR), age-related macular degeneration (AMD), and glaucoma. Since the retina is considered an extension of the brain, its unique structure allows it to exhibit similar immune response mechanisms to the brain. We further extend our exploration from the retina to the brain, examining the role of DNA methylation in brain development and its associated diseases, such as Alzheimer's disease (AD) and Huntington's disease (HD) to better understand the mechanistic links between retinal and brain diseases, and explore the possibility of communication between the visual system and the central nervous system (CNS) from an epigenetic perspective. Additionally, we discuss neurodevelopmental brain diseases, including schizophrenia (SZ), autism spectrum disorder (ASD), and intellectual disability (ID), focus on how DNA methylation affects neuronal development, synaptic plasticity, and cognitive function, providing insights into the molecular mechanisms underlying neurodevelopmental disorders.
Collapse
Affiliation(s)
- Chunxiu Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xuefei Fu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Toader C, Tataru CP, Munteanu O, Serban M, Covache-Busuioc RA, Ciurea AV, Enyedi M. Decoding Neurodegeneration: A Review of Molecular Mechanisms and Therapeutic Advances in Alzheimer's, Parkinson's, and ALS. Int J Mol Sci 2024; 25:12613. [PMID: 39684324 PMCID: PMC11641752 DOI: 10.3390/ijms252312613] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's, Parkinson's, ALS, and Huntington's, remain formidable challenges in medicine, with their relentless progression and limited therapeutic options. These diseases arise from a web of molecular disturbances-misfolded proteins, chronic neuroinflammation, mitochondrial dysfunction, and genetic mutations-that slowly dismantle neuronal integrity. Yet, recent scientific breakthroughs are opening new paths to intervene in these once-intractable conditions. This review synthesizes the latest insights into the underlying molecular dynamics of neurodegeneration, revealing how intertwined pathways drive the course of these diseases. With an eye on the most promising advances, we explore innovative therapies emerging from cutting-edge research: nanotechnology-based drug delivery systems capable of navigating the blood-brain barrier, gene-editing tools like CRISPR designed to correct harmful genetic variants, and stem cell strategies that not only replace lost neurons but foster neuroprotective environments. Pharmacogenomics is reshaping treatment personalization, enabling tailored therapies that align with individual genetic profiles, while molecular diagnostics and biomarkers are ushering in an era of early, precise disease detection. Furthermore, novel perspectives on the gut-brain axis are sparking interest as mounting evidence suggests that microbiome modulation may play a role in reducing neuroinflammatory responses linked to neurodegenerative progression. Taken together, these advances signal a shift toward a comprehensive, personalized approach that could transform neurodegenerative care. By integrating molecular insights and innovative therapeutic techniques, this review offers a forward-looking perspective on a future where treatments aim not just to manage symptoms but to fundamentally alter disease progression, presenting renewed hope for improved patient outcomes.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (M.S.); (R.-A.C.-B.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Calin Petru Tataru
- Ophthalmology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Octavian Munteanu
- Department of Anatomy, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Matei Serban
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (M.S.); (R.-A.C.-B.); (A.V.C.)
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (M.S.); (R.-A.C.-B.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (M.S.); (R.-A.C.-B.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
- Medical Section within the Romanian Academy, 010071 Bucharest, Romania
| | - Mihaly Enyedi
- Department of Anatomy, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
46
|
Bhat AA, Moglad E, Goyal A, Afzal M, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Ali H, Gaur A, Singh TG, Singh SK, Dua K, Gupta G. Nrf2 pathways in neuroprotection: Alleviating mitochondrial dysfunction and cognitive impairment in aging. Life Sci 2024; 357:123056. [PMID: 39277133 DOI: 10.1016/j.lfs.2024.123056] [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: 06/14/2024] [Revised: 08/27/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Mitochondrial dysfunction and cognitive impairment are widespread phenomena among the elderly, being crucial factors that contribute to neurodegenerative diseases. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important regulator of cellular defense systems, including that against oxidative stress. As such, increased Nrf2 activity may serve as a strategy to avert mitochondrial dysfunction and cognitive decline. Scientific data on Nrf2-mediated neuroprotection was collected from PubMed, Google Scholar, and Science Direct, specifically addressing mitochondrial dysfunction and cognitive impairment in older people. Search terms included "Nrf2", "mitochondrial dysfunction," "cognitive impairment," and "neuroprotection." Studies focusing on in vitro and in vivo models and clinical investigations were included to review Nrf2's therapeutic potential comprehensively. The relative studies have demonstrated that increased Nrf2 activity could improve mitochondrial performance, decrease oxidative pressure, and mitigate cognitive impairment. To a large extent, this is achieved through the modulation of critical cellular signalling pathways such as the Keap1/Nrf2 pathway, mitochondrial biogenesis, and neuroinflammatory responses. The present review summarizes the recent progress in comprehending the molecular mechanisms regarding the neuroprotective benefits mediated by Nrf2 through its substantial role against mitochondrial dysfunction and cognitive impairment. This review also emphasizes Nrf2-target pathways and their contribution to cognitive function improvement and rescue from mitochondria-related abnormalities as treatment strategies for neurodegenerative diseases that often affect elderly individuals.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589 Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341 Sakaka, Al-Jouf, Saudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Ashish Gaur
- Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India; Graphic Era Hill University, Clement Town, Dehradun 248002, India
| | | | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
47
|
Lo CH, Chen S, Zhao J, Liu Z, Wang B, Wang Q, Kowal TJ, Sun Y. Defective Neurogenesis in Lowe Syndrome is Caused by Mitochondria Loss and Cilia-related Sonic Hedgehog Defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621496. [PMID: 39553960 PMCID: PMC11565974 DOI: 10.1101/2024.11.01.621496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Human brain development is a complex process that requires intricate coordination of multiple cellular and developmental events. Dysfunction of lipid metabolism can lead to neurodevelopmental disorders. Lowe syndrome (LS) is a recessive X-linked disorder associated with proximal tubular renal disease, congenital cataracts and glaucoma, and central nervous system developmental delays. Mutations in OCRL, which encodes an inositol polyphosphate 5-phosphatase, lead to the development of LS. The cellular mechanism responsible for neuronal dysfunction in LS is unknown. Here we show depletion of mitochondrial DNA and decrease in mitochondrial activities result in neuronal differentiation defects. Increased astrocytes, which are secondary responders to neurodegeneration, are observed in neuronal (iN) cells differentiated from Lowe patient-derived iPSCs and an LS mouse model. Inactivation of cilia-related sonic hedgehog signaling, which organizes the pattern of cellular neuronal differentiation, is observed in an OCRL knockout, iN cells differentiated from Lowe patient-derived iPSCs, and an LS mouse model. Taken together, our findings indicate that mitochondrial dysfunction and impairment of the ciliary sonic hedgehog signaling pathway represent a novel pathogenic mechanism underlying the disrupted neuronal differentiation observed in LS.
Collapse
Affiliation(s)
- Chien-Hui Lo
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Siyu Chen
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jingyu Zhao
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Zhiquan Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Biao Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Qing Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tia J. Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
- Palo Alto Veterans Administration, Palo Alto, CA, USA
- Maternal Children Health Research Institute at Stanford, Stanford University School of Medicine, Palo Alto, CA, USA
- BioX, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
48
|
Cheng Y, Hussain SA, Alrubie TM, Zhang X. Neuroprotective Role of Transchalcone in Parkinson's Disease through AMP-activated Protein Kinase-mediated Signaling Pathway. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:312-320. [PMID: 39641138 DOI: 10.4103/ejpi.ejpi-d-24-00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
ABSTRACT Parkinson's disease (PD) is a gradually worsening neurodegenerative condition marked by the deterioration of dopaminergic neurons, motor dysfunction, and mitochondrial dysfunction. Trans-chalcone, a natural flavonoid, has shown promise in various disease models because of its antioxidant and anti-inflammatory features. This study investigates the neuroprotective effects of transchalcone in a rat model of PD, focusing on its impact on the activation levels of AMP-activated protein kinase (AMPK) signaling pathway, sirtuin1 (SIRT1) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) proteins, and mitochondrial-inflammatory responses. Male Sprague Dawley rats were allocated into five groups Control, Control plus transchalcone, PD, PD plus transchalcone, PD plus compound-C, and PD plus Compound-C and trans-chalcone. PD was induced using intranigral 6-hydroxydopamine injection. Trans-chalcone (100 μg/kg) and compound-C (20 mg/kg) were intraperitoneally administered daily for 4 weeks in PD rats. Motor function was assessed using rota-rod and grid tests. Striatal dopamine and cytokines (interleukin 1-beta [IL-1 β], IL-10) and p65-nuclear factor kappa-B (NF-κB) levels were measured with enzyme-linked immunosorbent assay. Mitochondrial function was evaluated by fluorometric techniques. The expression of phosphorylated AMPK, PGC-1α, and SIRT1 was analyzed by Western blotting. Trans-chalcone treatment significantly improved motor function, evidenced by increased latency to fall in the rota-rod test and recovered traversal time in the grid test. It also restored dopamine levels, enhanced mitochondrial function (reduced reactive oxygen species levels, increased membrane potential, and adenosine triphosphate production), normalized cytokines (IL-1 β, IL-10) and p65-NF-κB, and upregulated the proteins expression in rats with PD. Inhibition of AMPK activity with compound-C suppressed the neuroprotective impacts of trans-chalcone, highlighting the contribution of AMPK signaling pathway in its mechanism of action. Neuroprotective and mitoprotective impacts of trans-chalcone were mostly mediated through the activation of AMPK-SIRT1-PGC1α pathway. These results indicate that trans-chalcone could be a promising therapeutic agent for PD, warranting further investigation to assess its efficacy and safety in human patients.
Collapse
Affiliation(s)
- Yao Cheng
- Department of Neurology, Harbin 242 Hospital, Harbin, China
| | - Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Turki Mayudh Alrubie
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Xiaomin Zhang
- Department of Neurology, The Affiliated Hospital of Yunnan University, Kunming, China
| |
Collapse
|
49
|
Kazemeini S, Nadeem-Tariq A, Shih R, Rafanan J, Ghani N, Vida TA. From Plaques to Pathways in Alzheimer's Disease: The Mitochondrial-Neurovascular-Metabolic Hypothesis. Int J Mol Sci 2024; 25:11720. [PMID: 39519272 PMCID: PMC11546801 DOI: 10.3390/ijms252111720] [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: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) presents a public health challenge due to its progressive neurodegeneration, cognitive decline, and memory loss. The amyloid cascade hypothesis, which postulates that the accumulation of amyloid-beta (Aβ) peptides initiates a cascade leading to AD, has dominated research and therapeutic strategies. The failure of recent Aβ-targeted therapies to yield conclusive benefits necessitates further exploration of AD pathology. This review proposes the Mitochondrial-Neurovascular-Metabolic (MNM) hypothesis, which integrates mitochondrial dysfunction, impaired neurovascular regulation, and systemic metabolic disturbances as interrelated contributors to AD pathogenesis. Mitochondrial dysfunction, a hallmark of AD, leads to oxidative stress and bioenergetic failure. Concurrently, the breakdown of the blood-brain barrier (BBB) and impaired cerebral blood flow, which characterize neurovascular dysregulation, accelerate neurodegeneration. Metabolic disturbances such as glucose hypometabolism and insulin resistance further impair neuronal function and survival. This hypothesis highlights the interconnectedness of these pathways and suggests that therapeutic strategies targeting mitochondrial health, neurovascular integrity, and metabolic regulation may offer more effective interventions. The MNM hypothesis addresses these multifaceted aspects of AD, providing a comprehensive framework for understanding disease progression and developing novel therapeutic approaches. This approach paves the way for developing innovative therapeutic strategies that could significantly improve outcomes for millions affected worldwide.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (S.K.); (A.N.-T.); (R.S.); (J.R.); (N.G.)
| |
Collapse
|
50
|
Loers G, Bork U, Schachner M. Functional Relationships between L1CAM, LC3, ATG12, and Aβ. Int J Mol Sci 2024; 25:10829. [PMID: 39409157 PMCID: PMC11476435 DOI: 10.3390/ijms251910829] [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: 09/20/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
Abnormal protein accumulations in the brain are linked to aging and the pathogenesis of dementia of various types, including Alzheimer's disease. These accumulations can be reduced by cell indigenous mechanisms. Among these is autophagy, whereby proteins are transferred to lysosomes for degradation. Autophagic dysfunction hampers the elimination of pathogenic protein aggregations that contribute to cell death. We had observed that the adhesion molecule L1 interacts with microtubule-associated protein 1 light-chain 3 (LC3), which is needed for autophagy substrate selection. L1 increases cell survival in an LC3-dependent manner via its extracellular LC3 interacting region (LIR). L1 also interacts with Aβ and reduces the Aβ plaque load in an AD model mouse. Based on these results, we investigated whether L1 could contribute to autophagy of aggregated Aβ and its clearance. We here show that L1 interacts with autophagy-related protein 12 (ATG12) via its LIR domain, whereas interaction with ubiquitin-binding protein p62/SQSTM1 does not depend on LIR. Aβ, bound to L1, is carried to the autophagosome leading to Aβ elimination. Showing that the mitophagy-related L1-70 fragment is ubiquitinated, we expect that the p62/SQSTM1 pathway also contributes to Aβ elimination. We propose that enhancing L1 functions may contribute to therapy in humans.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ute Bork
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| |
Collapse
|