1
|
Parchwani D, Singh R, Patel D. Biological and translational attributes of mitochondrial DNA copy number: Laboratory perspective to clinical relevance. World J Methodol 2025; 15:102709. [DOI: 10.5662/wjm.v15.i3.102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/21/2025] [Accepted: 02/08/2025] [Indexed: 03/06/2025] Open
Abstract
The mitochondrial DNA copy number (mtDNAcn) plays a vital role in cellular energy metabolism and mitochondrial health. As mitochondria are responsible for adenosine triphosphate production through oxidative phosphorylation, maintaining an appropriate mtDNAcn level is vital for the overall cellular function. Alterations in mtDNAcn have been linked to various diseases, including neurodegenerative disorders, metabolic conditions, and cancers, making it an important biomarker for understanding the disease pathogenesis. The accurate estimation of mtDNAcn is essential for clinical applications. Quantitative polymerase chain reaction and next-generation sequencing are commonly employed techniques with distinct advantages and limitations. Clinically, mtDNAcn serves as a valuable indicator for early diagnosis, disease progression, and treatment response. For instance, in oncology, elevated mtDNAcn levels in blood samples are associated with tumor aggressiveness and can aid in monitoring treatment efficacy. In neurodegenerative diseases such as Alzheimer’s and Parkinson’s, altered mtDNAcn patterns provide insights into disease mechanisms and progression. Understanding and estimating mtDNAcn are critical for advancing diagnostic and therapeutic strategies in various medical fields. As research continues to uncover the implications of mtDNAcn alterations, its potential as a clinical biomarker is likely to expand, thereby enhancing our ability to diagnose and manage complex diseases.
Collapse
Affiliation(s)
- Deepak Parchwani
- Department of Biochemistry, All India Institute of Medical Sciences, Rajkot 360001, India
| | - Ragini Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Rajkot 360001, India
| | - Digisha Patel
- Department of Physiology, Shantabaa Medical College and General Hospital Amreli, Amreli 365601, Gujarāt, India
| |
Collapse
|
2
|
Pradeepkiran JA, Islam MA, Sehar U, Reddy AP, Vijayan M, Reddy PH. Impact of diet and exercise on mitochondrial quality and mitophagy in Alzheimer's disease. Ageing Res Rev 2025; 108:102734. [PMID: 40120948 DOI: 10.1016/j.arr.2025.102734] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/26/2024] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that affects millions of people worldwide. It is characterized by the accumulation of beta-amyloid and phosphorylated tau, synaptic damage, and mitochondrial abnormalities in the brain, leading to the progressive loss of cognitive function and memory. In AD, emerging research suggests that lifestyle factors such as a healthy diet and regular exercise may play a significant role in delaying the onset and progression of the disease. Mitochondria are often referred to as the powerhouse of the cell, as they are responsible for producing the energy to cells, including neurons to maintain cognitive function. Our article elaborates on how mitochondrial quality and function decline with age and AD, leading to an increase in oxidative stress and a decrease in ATP production. Decline in mitochondrial quality can impair cellular functions contributing to the development and progression of disease with the loss of neuronal functions in AD. This article also covered mitophagy, the process by which damaged or dysfunctional mitochondria are selectively removed from the cell to maintain cellular homeostasis. Impaired mitophagy has been implicated in the progression and pathogenesis of AD. We also discussed the impact of impaired mitophagy implicated in AD, as the accumulation of damaged mitochondria can lead to increased oxidative stress. We expounded how dietary interventions and exercise can help to improve mitochondrial quality, and mitochondrial function and enhance mitophagy in AD. A diet rich in antioxidants, polyphenols, and mitochondria-targeted small molecules has been shown to enhance mitochondrial function and protect against oxidative stress, particularly in neurons with aged and mild cognitively impaired subjects and AD patients. Promoting a healthy lifestyle, mainly balanced diet and regular exercise that support mitochondrial health, in an individual can potentially delay the onset and progression of AD. In conclusion, a healthy diet and regular exercise play a crucial role in maintaining mitochondrial quality and mitochondrial function, in turn, enhancing mitophagy and synaptic activities that delay AD in the elderly populations.
Collapse
Affiliation(s)
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
3
|
Nambidi S, Pallatt S, Banerjee A, Pathak S, Chan MKS. Klotho protein: a multifaceted regulator in aging and cancer dynamics. Mol Biol Rep 2025; 52:507. [PMID: 40423846 DOI: 10.1007/s11033-025-10575-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 05/04/2025] [Indexed: 05/28/2025]
Abstract
Klotho, named after the youngest of the three Fates in Greek mythology daughters of Zeus and Nyx, who together spin the thread of life, allot destiny, and determine the time of passing for both mortals and immortals, is an important regulatory factor in aging and cancer dynamics. Initially described as an aging-suppressing protein, Klotho is now recognized for its more diverse role in modulating key signaling pathways like Wnt/β-catenin, IGF-1, PI3K/AKT, and TGF-β. Essentially, its various pro-cellular health functions, such as antioxidant, anti-inflammatory, and tumor-suppressive activities, are, in fact, considered that ensures the maintenance of cellular health and reduce complications related to aging. Klotho deficiency is associated with accelerated aging, chronic kidney disease, cardiovascular disorders, neurodegeneration, and various cancers. This review thus covers the twin roles of Klotho as an antiaging and tumor-suppressor protein, on their therapeutic potential, as well as advances in delivery systems and development of biomarkers and challenges for clinical translation.. Moreover, natural strategies like exercise and dietary interventions are explored that could help overcome Klotho deficiency. Further research with Klotho may offer a paradigm shift in the treatment of aging and cancer and add yet another avenue to increase survival of the patients.
Collapse
Affiliation(s)
- Sibin Nambidi
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, Tamil Nadu, India
| | - Sneha Pallatt
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, Tamil Nadu, India
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, Tamil Nadu, India.
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, Tamil Nadu, India.
| | - Mike K S Chan
- European Wellness Biomedical Group, Klosterstrasse 205, 67480, Edenkoben, Germany
- Baden R&D Laboratories GmbH, Ferdinand-Lassalle-Strasse 40, 72770, Reutlingen, Germany
| |
Collapse
|
4
|
Mashayekhi-Sardoo H, Razazpour F, Hakemi Z, Hedayati-Moghadam M, Baghcheghi Y. Ethanol-Induced Depression: Exploring the Underlying Molecular Mechanisms. Cell Mol Neurobiol 2025; 45:49. [PMID: 40405002 PMCID: PMC12098258 DOI: 10.1007/s10571-025-01569-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/12/2025] [Indexed: 05/24/2025]
Abstract
Ethanol consumption is widely recognized for its detrimental effects on mental health, particularly its association with depressive disorders. This narrative review aims to explore the intricate molecular mechanisms underlying ethanol-induced depression, synthesizing findings from preclinical and clinical studies. We begin by providing an overview of the relationship between chronic ethanol consumption and depression, highlighting compelling evidence from diverse populations. Subsequently, we delve into insights from animal models that elucidate the pathophysiological changes triggered by prolonged ethanol exposure. Key mechanisms identified include oxidative stress, which contributes to cellular damage; neuroinflammation, characterized by the activation of glial cells and altered cytokine profiles; and disruptions in neurotrophic factors that impair neuronal growth and survival. Furthermore, we discuss the induction of apoptosis in neural cells and the significant impact of ethanol on neurotransmitter receptor remodeling and regulation, leading to altered synaptic transmission. While substantial progress has been made in understanding these mechanisms, we also acknowledge the limitations of current research methodologies and call for further investigations to translate these findings into effective therapeutic strategies for individuals affected by ethanol-induced depression. This review ultimately underscores the need for a comprehensive understanding of the molecular underpinnings of ethanol's impact on mood disorders, paving the way for improved interventions and preventative measures.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-Sardoo
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran
- School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fateme Razazpour
- Oral and Dental Diseases Research Center, Kerman University of Medical Science, Kerman, Iran
| | - Zohreh Hakemi
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Mahdiyeh Hedayati-Moghadam
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, 7861755765, Iran.
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
5
|
Dar NJ, Currais A, Taguchi T, Andrews N, Maher P. Cannabinol (CBN) alleviates age-related cognitive decline by improving synaptic and mitochondrial health. Redox Biol 2025; 84:103692. [PMID: 40412024 DOI: 10.1016/j.redox.2025.103692] [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: 02/21/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Age-related cognitive decline and neurodegenerative diseases, such as Alzheimer's disease, represent major global health challenges, particularly with an aging population. Mitochondrial dysfunction appears to play a central role in the pathophysiology of these conditions by driving redox dysregulation and impairing cellular energy metabolism. Despite extensive research, effective therapeutic options remain limited. Cannabinol (CBN), a cannabinoid previously identified as a potent inhibitor of oxytosis/ferroptosis through mitochondrial modulation, has demonstrated promising neuroprotective effects. In cell culture, CBN targets mitochondria, preserving mitochondrial membrane potential, enhancing antioxidant defenses and regulating bioenergetic processes. However, the in vivo therapeutic potential of CBN, particularly in aging models, has not been thoroughly explored. To address this gap, this study investigated the effects of CBN on age-associated cognitive decline and metabolic dysfunction using the SAMP8 mouse model of accelerated aging. Our results show that CBN significantly improves spatial learning and memory, with more pronounced cognitive benefits observed in female mice. These cognitive improvements are accompanied by sex-specific changes in metabolic parameters, such as enhanced oxygen consumption and energy expenditure. Mechanistically, CBN modulates key regulators of mitochondrial dynamics, including mitofusin 2 (MFN2) and dynamin-related protein 1 (DRP1), while upregulating markers of mitochondrial biogenesis including mitochondrial transcription factor A (TFAM) and translocase of outer mitochondrial membrane 20 (TOM20). Additionally, CBN upregulates key synaptic proteins involved in vesicle trafficking and postsynaptic signaling suggesting that it enhances synaptic function and neurotransmission, further reinforcing its neuroprotective effects. This study provides in vivo evidence supporting CBN's potential to mitigate age-related cognitive and metabolic dysfunction, with notable sex-specific effects, highlighting its promise for neurodegenerative diseases and cognitive decline.
Collapse
Affiliation(s)
- Nawab John Dar
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Antonio Currais
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Taketo Taguchi
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nick Andrews
- Behaviour Testing Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Pamela Maher
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
6
|
Dang M, Wu L, Zhang X. Structural insights and milestones in TDP-43 research: A comprehensive review of its pathological and therapeutic advances. Int J Biol Macromol 2025; 306:141677. [PMID: 40032118 DOI: 10.1016/j.ijbiomac.2025.141677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Transactive response (TAR) DNA-binding protein 43 (TDP-43) is a critical RNA/DNA-binding protein involved in various cellular processes, including RNA splicing, transcription regulation, and RNA stability. Mislocalization and aggregation of TDP-43 in the cytoplasm are key features of the pathogenesis of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Alzheimer's disease (AD). This review provides a comprehensive retrospective and prospective analysis of TDP-43 research, highlighting structural insights, significant milestones, and the evolving understanding of its physiological and pathological functions. We delineate five major stages in TDP-43 research, from its initial discovery as a pathological hallmark in neurodegeneration to the recent advances in understanding its liquid-liquid phase separation (LLPS) behavior and interactions with cellular processes. Furthermore, we assess therapeutic strategies targeting TDP-43 pathology, categorizing approaches into direct and indirect interventions, alongside modulating aberrant TDP-43 LLPS. We propose that future research will focus on three critical areas: targeting TDP-43 structural polymorphisms for disease-specific therapeutics, exploring dual temporal-spatial modulation of TDP-43, and advancing nano-therapy. More importantly, we emphasize the importance of understanding TDP-43's functional repertoire at the mesoscale, which bridges its molecular functions with broader cellular processes. This review offers a foundational framework for advancing TDP-43 research and therapeutic development.
Collapse
Affiliation(s)
- Mei Dang
- Chinese-German Joint Institute for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China; Department of Biological Sciences, Faculty of Science, National University of Singapore, 10 Kent Ridge Crescent, 119260, Singapore
| | - Longjiang Wu
- Chinese-German Joint Institute for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China
| | - Xiaoying Zhang
- Chinese-German Joint Institute for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, China; Centre of Molecular & Environmental Biology, Department of Biology, University of Minho, 4710-057 Braga, Portugal; Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, N1G 2W1 Guelph, Ontario, Canada.
| |
Collapse
|
7
|
Ye J, Dai X, Zhang C, Duan Z, Zhou G, Wang J. Investigating the causal relationships between mitochondrial proteins and dementia with Lewy bodies. J Alzheimers Dis 2025; 105:378-386. [PMID: 40111912 DOI: 10.1177/13872877251328882] [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: 03/22/2025]
Abstract
BackgroundDisruptions in mitochondrial function have been implicated in various neurodegenerative diseases. However, the specific role of mitochondrial proteins in the pathogenesis of dementia with Lewy bodies (DLB) remains poorly understood.ObjectiveThis study aims to investigate potential causal relationships between mitochondrial proteins and DLB risk using Mendelian randomization (MR) analysis.MethodsCausal associations between 66 mitochondrial proteins (MPs) and DLB were assessed by MR analysis, utilizing data from comprehensive genome-wide association studies (GWAS), with various analytical methods, including the inverse variance weighted, MR-Egger, and weighted median. Cochran's Q statistics assessed the heterogeneity of instrumental variables.ResultsGenetic predispositions to increased levels of ES1 protein homolog and apoptosis-inducing factor 1 (AIF-1) were associated with an elevated risk of DLB. Conversely, genetic predispositions to increased levels of glutaredoxin-2 (GLRX-2), complement component 1 Q subcomponent-binding protein (C1QBP), and mitochondrial glutamate carrier 2 (GC2) were found to be protective against DLB. Sensitivity analyses revealed no heterogeneity or horizontal pleiotropy among the selected instrumental variables.ConclusionsOur MR study identifies specific MPs potentially causally linked to DLB risk. These findings offer new insights into the MP-related mechanisms underlying DLB pathogenesis and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Jingna Ye
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Xuelian Dai
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Canwen Zhang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Zhihui Duan
- Department of Neurology, Luoyang Center Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Guoqing Zhou
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Juan Wang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| |
Collapse
|
8
|
Juhász L, Spisák K, Szolnoki BZ, Nászai A, Szabó Á, Rutai A, Tallósy SP, Szabó A, Toldi J, Tanaka M, Takeda K, Ozaki K, Inoue H, Yamamoto S, Ono E, Boros M, Kaszaki J, Vécsei L. The Power Struggle: Kynurenine Pathway Enzyme Knockouts and Brain Mitochondrial Respiration. J Neurochem 2025; 169:e70075. [PMID: 40317489 PMCID: PMC12048769 DOI: 10.1111/jnc.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/21/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025]
Abstract
Numerous illnesses, including neurological and mental disorders, have been associated with mitochondrial dysfunction. Disruptions in mitochondrial respiration and energy production have been linked to dysmetabolism of the tryptophan (Trp)-kynurenine (KYN) pathway, which produces a diverse array of bioactive metabolites. Kynurenic acid (KYNA) is a putative neuroprotectant. The exact mechanisms through which Trp-KYN metabolic dysregulation affects mitochondrial function remain largely unclear. This study investigates the impact of the genetic deletion of kynurenine aminotransferase (KAT) enzymes, which are responsible for KYNA synthesis, on mitochondrial function, specifically mitochondrial respiration and ATP synthesis, and its potential role in neuropsychiatric pathology. CRISPR/Cas9-induced knockout mouse strains kat1-/-, kat2-/-, and kat3-/- were generated. Eight-to-ten-week-old male mice were used, and cerebral and hepatic respiration, complex I- and II-linked oxidative phosphorylation (CI and CII OXPHOS), and complex IV (CIV) activity were measured using high-resolution respirometry. Mitochondrial membrane potential changes were measured with Fluorescence-Sensor Blue and safranin dye. KAT knockout mice exhibited significantly lower cerebellar respiration (CI OXPHOS, CII OXPHOS, and CIV activity) compared to wild-type mice. Lower baseline respiration and attenuated OXPHOS activities were observed in the hippocampus and striatum, particularly in kat2-/- and kat3-/- mice. Non-neuronal tissues showed reduced CIV activity, while ADP-stimulated CI and CII OXPHOS remained unchanged. The deletion of the KAT genes significantly impairs mitochondrial respiration and ATP synthesis, potentially contributing to pathogenesis. This study highlights the importance of KYNA in mitochondrial function, offering new insights into potential therapeutic targets for various disorders. Targeting the KYN pathway could mitigate mitochondrial dysfunction in a variety of diseased conditions.
Collapse
Affiliation(s)
- László Juhász
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - Krisztina Spisák
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - Boglárka Zsuzsa Szolnoki
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - Anna Nászai
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - Ágnes Szabó
- Department of NeurologyUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
- HUN‐REN‐SZTE Neuroscience Research Group, Hungarian Research NetworkUniversity of Szeged (HUN‐REN‐SZTE), Danube Neuroscience Research LaboratorySzegedHungary
| | - Attila Rutai
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - Szabolcs Péter Tallósy
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - Andrea Szabó
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - József Toldi
- Department of Physiology, Anatomy and NeuroscienceUniversity of SzegedSzegedHungary
| | - Masaru Tanaka
- HUN‐REN‐SZTE Neuroscience Research Group, Hungarian Research NetworkUniversity of Szeged (HUN‐REN‐SZTE), Danube Neuroscience Research LaboratorySzegedHungary
| | - Keiko Takeda
- Department of Biomedicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kinuyo Ozaki
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Hiromi Inoue
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Sayo Yamamoto
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Etsuro Ono
- Department of Biomedicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Mihály Boros
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - József Kaszaki
- Institute of Surgical ResearchUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
| | - László Vécsei
- Department of NeurologyUniversity of Szeged, Albert Szent‐Györgyi Medical SchoolSzegedHungary
- HUN‐REN‐SZTE Neuroscience Research Group, Hungarian Research NetworkUniversity of Szeged (HUN‐REN‐SZTE), Danube Neuroscience Research LaboratorySzegedHungary
| |
Collapse
|
9
|
Umapathy S, Pan I. Glucose reduced nano-Se mitigates Cu-induced ROS by upregulating antioxidant genes in zebrafish larvae. NANOSCALE ADVANCES 2025; 7:2502-2517. [PMID: 40061839 PMCID: PMC11887129 DOI: 10.1039/d4na00644e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 02/21/2025] [Indexed: 05/01/2025]
Abstract
This study compares the therapeutic efficiency of bovine serum albumin-stabilized selenium nanoparticles in reducing oxidative stress and improving cellular health. The nanoparticles were synthesized using mussel-extracted selenium with two reducing agents: d-glucose and orange. Inductively coupled plasma-optical emission spectroscopy and X-ray diffraction analyses confirmed the presence of selenium. The reducing agent and duration influenced the nanoparticle size. Reduction with d-glucose for 1 hour revealed that the particles exhibited an average size of 10 nm. Copper sulfate-induced malformations such as yolk sac and pericardial edema were observed with 25 μg ml-1 of orange-reduced nanoparticles, while d-glucose-reduced nanoparticles mitigated these malformations at 25 μg ml-1. Treatment with stabilized Se-NPs reduced with d-glucose for 30 minutes showed 33% dose-dependent radical scavenging activities, upregulated approximately 2-fold of superoxide dismutase, catalase, glutathione reductase, and glutathione peroxidase encoding genes and restored homeostasis by decreasing lipid peroxidation (27.32 nmol mg-1 ml-1) and nitric oxide levels (6.71 μM). They also had the potential to restore cognitive properties such as larval movement (93.40 m) without altering larval behaviour. Live cell imaging indicated a significant decrease in cellular reactive oxygen species and lipid peroxidation levels in the gut and liver. These findings suggest that Se-NPs reduced for 30 minutes with d-glucose are promising candidates for oxidative stress-induced neurodegeneration.
Collapse
Affiliation(s)
- Suganiya Umapathy
- Institute of Biotechnology, Department of Medical Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences Thandalam Chennai 602 105 Tamil Nadu India
| | - Ieshita Pan
- Institute of Biotechnology, Department of Medical Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences Thandalam Chennai 602 105 Tamil Nadu India
| |
Collapse
|
10
|
Adlimoghaddam A, Fayazbakhsh F, Mohammadi M, Babaei Z, Behrooz AB, Tabasi F, Guan T, Beheshti I, Aghaei M, Klionsky DJ, Albensi BC, Ghavami S. Sex and region-specific disruption of autophagy and mitophagy in Alzheimer's disease: linking cellular dysfunction to cognitive decline. Cell Death Discov 2025; 11:204. [PMID: 40287423 PMCID: PMC12033262 DOI: 10.1038/s41420-025-02490-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Macroautophagy and mitophagy are critical processes in Alzheimer's disease (AD), yet their links to behavioral outcomes, particularly sex-specific differences, are not fully understood. This study investigates autophagic (LC3B-II, SQSTM1) and mitophagic (BNIP3L, BNIP3, BCL2L13) markers in the cortex and hippocampus of male and female 3xTg-AD mice, using western blotting, transmission electron microscopy (TEM), and behavioral tests (novel object recognition and novel object placement). Significant sex-specific differences emerged: female 3xTg-AD mice exhibited autophagosome accumulation due to impaired degradation in the cortex, while males showed fewer autophagosomes, especially in the hippocampus, without significant degradation changes. TEM analyses demonstrated variations in mitochondrial and mitophagosome numbers correlated with memory outcomes. Females had enhanced mitophagy, with higher BNIP3L and BCL2L13 levels, whereas males showed elevated BNIP3 dimers. Cognitive deficits in females correlated with mitochondrial dysfunction in the cortex, while in males, higher LC3B-II levels associated positively with cognitive performance, suggesting protective autophagy effects. Using machine learning, we predicted mitophagosome and mitochondrial numbers based on behavioral data, pioneering a predictive approach to cellular outcomes in AD. These findings underscore the importance of sex-specific regulation of autophagy and mitophagy in AD and support personalized therapeutic approaches targeting these pathways. Integrating machine learning emphasizes its potential to advance neurodegenerative research. Sex-specific differences in autophagy and mitophagy regulation in Alzheimer's disease (AD) are highlighted. Female 3xTg-AD mice show autophagosome accumulation and cognitive deficits, while males exhibit variations in mitophagy markers and behavior.
Collapse
Affiliation(s)
- Aida Adlimoghaddam
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Autophagy Research Centre, SUMS, Shiraz, Fars, Iran
| | - Fariba Fayazbakhsh
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Mohsen Mohammadi
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Zeinab Babaei
- Department of Clinical Biochemistry and Biophysics, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Farhad Tabasi
- Department of Neurosurgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Teng Guan
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Iman Beheshti
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Mahmoud Aghaei
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Daniel J Klionsky
- Department of Molecular, Cellular and Developmental Biology and Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Benedict C Albensi
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL, USA.
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada.
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada.
- St. Boniface Hospital Research, Winnipeg, MB, Canada.
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555, Katowice, Poland.
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
11
|
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
|
12
|
Gerardo H, Lourenço T, Torres J, Ferreira M, Aveleira C, Simões S, Ferreira L, Cavadas C, Oliveira PJ, Teixeira J, Grãos M. Extracellular matrix mechanical cues (dys)regulate metabolic redox homeostasis due to impaired autophagic flux. Eur J Clin Invest 2025:e70051. [PMID: 40280877 DOI: 10.1111/eci.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Extracellular matrix (ECM) stiffness is increasingly recognized as a critical regulator of cellular behaviour, governing processes such as proliferation, differentiation, and metabolism. Neurodegenerative diseases are characterized by mitochondrial dysfunction, oxidative stress, impaired autophagy, and progressive softening of the brain tissue, yet research into how mechanical cues influence cellular metabolism in this context remains scarce. MATERIALS AND METHODS In this study, we evaluated the long-term effects of brain-compliant, soft ECM on mitochondrial bioenergetics, redox balance, and autophagic capacity in human neuroblastoma (SH-SY5Y) and mouse hippocampal (HT22) cell lines, as well as primary mouse neurons. RESULTS We observed that prolonged exposure to soft ECM does not impact cell proliferative capacity of neuronal cells but results in mitochondrial bioenergetic dysfunction, redox imbalance, and disrupted autophagic flux. These findings were consistently validated across both human and mouse neuronal cells. Our data indicate a decreased maximal autophagic capacity in cells exposed to long-term soft ECM, potentially due to an imbalance in autophagosome formation and degradation, as demonstrated by decreased LC3 II levels following chloroquine-induced autophagic flux inhibition. This impairment in autophagy was coupled with increased cellular oxidative stress, further indicating metabolic alterations. CONCLUSIONS These findings emphasize the critical role of ECM stiffness in regulating neuronal cell metabolism and suggest that prolonged exposure to soft ECM may mimic key aspects of neurodegenerative disease pathology, thereby enhancing the physiological relevance of in vitro models. This study underscores the necessity for further research into ECM mechanics as a contributing factor in neurodegenerative disease progression and as a potential target for therapeutic strategies.
Collapse
Affiliation(s)
- Heloísa Gerardo
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Tânia Lourenço
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Júlio Torres
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research PDBEB - Doctoral Programme in Experimental Biology and Biomedicine, University of Coimbra, Coimbra, Portugal
| | - Manuela Ferreira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Célia Aveleira
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Aging - MIA, Coimbra, Portugal
| | - Susana Simões
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - José Teixeira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Department of Life Science (DCV), Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | - Mário Grãos
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
13
|
Basak B, Holzbaur ELF. Mitophagy in Neurons: Mechanisms Regulating Mitochondrial Turnover and Neuronal Homeostasis. J Mol Biol 2025:169161. [PMID: 40268233 DOI: 10.1016/j.jmb.2025.169161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
Mitochondrial quality control is instrumental in regulating neuronal health and survival. The receptor-mediated clearance of damaged mitochondria by autophagy, known as mitophagy, plays a key role in controlling mitochondrial homeostasis. Mutations in genes that regulate mitophagy are causative for familial forms of neurological disorders including Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS). PINK1/Parkin-dependent mitophagy is the best studied mitophagy pathway, while more recent work has brought to light additional mitochondrial quality control mechanisms that operate either in parallel to or independent of PINK1/Parkin mitophagy. Here, we discuss our current understanding of mitophagy mechanisms operating in neurons to govern mitochondrial homeostasis. We also summarize progress in our understanding of the links between mitophagic dysfunction and neurodegeneration, and highlight the potential for therapeutic interventions to maintain mitochondrial health and neuronal function.
Collapse
Affiliation(s)
- Bishal Basak
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
14
|
Jung YH, Jo HY, Kim DH, Oh YJ, Kim M, Na S, Song HY, Lee HJ. Exosome-Mediated Mitochondrial Regulation: A Promising Therapeutic Tool for Alzheimer's Disease and Parkinson's Disease. Int J Nanomedicine 2025; 20:4903-4917. [PMID: 40259919 PMCID: PMC12011032 DOI: 10.2147/ijn.s513816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are representative neurodegenerative diseases with abnormal energy metabolism and altered distribution and deformation of mitochondria within neurons, particularly in brain regions such as the hippocampus and substantia nigra. Neurons have high energy demands; thus, maintaining a healthy mitochondrial population is important for their biological function. Recently, exosomes have been reported to have mitochondrial regulatory potential and antineurodegenerative properties. This review presents the mitochondrial abnormalities in brain cells associated with AD and PD and the potential of exosomes to treat these diseases. Specifically, it recapitulates research on the molecular mechanisms whereby exosomes regulate mitochondrial biogenesis, fusion/fission dynamics, mitochondrial transport, and mitophagy. Furthermore, this review discusses exosome-triggered signaling pathways that regulate nuclear factor (erythroid-derived 2)-like 2-dependent mitochondrial antioxidation and hypoxia inducible factor 1α-dependent metabolic reprogramming. In summary, this review aims to provide a profound understanding of the regulatory effect of exosomes on mitochondrial function in neurons and to propose exosome-mediated mitochondrial regulation as a promising strategy for AD and PD.
Collapse
Affiliation(s)
- Young Hyun Jung
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Hyo Youn Jo
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Dae Hyun Kim
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Yeon Ju Oh
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Minsoo Kim
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Seunghyun Na
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| |
Collapse
|
15
|
Dehkordi SK, Sajedi S, Heshmat A, Orr ME, Zare H. Identification of markers for neurescence through transcriptomic profiling of postmortem human brains. RESEARCH SQUARE 2025:rs.3.rs-5903682. [PMID: 40297699 PMCID: PMC12036471 DOI: 10.21203/rs.3.rs-5903682/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Neuronal senescence (i.e., neurescent) is an important hallmark of aging and neurodegeneration, but it remains poorly characterized in the human brain due to the lack of reliable markers. This study aimed to identify neurescent markers based on single-nucleus transcriptome data from postmortem human prefrontal cortex. Using an eigengene approach, we integrated three gene panels: a) SenMayo, b) Canonical Senescence Pathway (CSP), and c) Senescence Initiating Pathway (SIP), to identify neurescent signatures. We found that paired markers outperform single markers; for instance, by combining CDKN2D and ETS2 in a decision tree, a high accuracy of 99% and perfect specificity (100%) were achieved in distinguishing neurescent. Differential expression analyses identified 324 genes that are overexpressed in neurescent. These genes showed significant associations with important neurodegeneration-related pathways including Alzheimer's disease, Parkinson's disease, and Huntington's disease. Interestingly, several of these overexpressed genes are linked to mitochondrial dysfunction and cytoskeletal dysregulation. These findings provide valuable insights into the complexities of neurescent, emphasizing the need for further exploration of histologically viable markers and validation in broader datasets.
Collapse
Affiliation(s)
| | | | | | | | - Habil Zare
- The University of Texas Health Science Center at San Antonio
| |
Collapse
|
16
|
Zuo X. Mitochondrial Imbalance in Down Syndrome: A Driver of Accelerated Brain Aging? Aging Dis 2025:AD.2025.0189. [PMID: 40249934 DOI: 10.14336/ad.2025.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/06/2025] [Indexed: 04/20/2025] Open
Abstract
Down syndrome (DS), caused by trisomy of chromosome 21 (HSA21), is a complex condition associated with neurodevelopmental impairments and accelerated brain aging, often culminating in early-onset Alzheimer's disease (AD). Central to this accelerated aging is mitochondrial imbalance, characterized by disrupted energy metabolism, increased oxidative stress, impaired dynamics, and defective quality control mechanisms like mitophagy. These abnormalities exacerbate neuronal vulnerability, driving cognitive decline and neurodegeneration. This review examines the genetic and biochemical underpinnings of mitochondrial dysfunction in DS, with a focus on the role of HSA21-encoded genes. We also highlight how mitochondrial dysfunction, amplified by oxidative stress and HSA21 gene dosage effects, converges with cellular senescence and neuroinflammation to accelerate Alzheimer-like pathology and brain aging in DS. Finally, we discuss emerging therapeutic strategies targeting mitochondrial pathways, which hold promise for mitigating neurodegenerative phenotypes and improving outcomes in DS.
Collapse
|
17
|
Ashwani, Sharma A, Choudhary MK, Gugulothu D, Pandita D, Verma S, Vora LK, Khatri DK, Garabadu D. Epigenetic and Mitochondrial Metabolic Dysfunction in Multiple Sclerosis: A Review of Herbal Drug Approaches and Current Clinical Trials. Mol Neurobiol 2025:10.1007/s12035-025-04868-8. [PMID: 40180689 DOI: 10.1007/s12035-025-04868-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disease characterised by inflammation, demyelination, and neurodegeneration within the central nervous system (CNS). While the exact causes remain unclear, recent research highlights the significant role of epigenetic modifications and mitochondrial dysfunction in the disease's onset and progression. Epigenetic alterations, such as DNA methylation, histone modification, and microRNA regulation, influence gene expression without altering the DNA sequence, leading to immune dysregulation and inflammation. Similarly, mitochondrial dysfunction, marked by impaired oxidative phosphorylation, reduced adenosine triphosphate (ATP) production, and increased reactive oxygen species (ROS), contributes to neurodegeneration and impaired remyelination in MS. The growing interest in targeting these two interconnected mechanisms has opened new avenues for MS treatment. Herbal drugs, known for their multi-targeted effects, have shown potential in modulating epigenetic markers and enhancing mitochondrial function. Compounds such as resveratrol, curcumin, epigallocatechin-3-gallate (EGCG), quercetin, and omega-3 fatty acids demonstrate potential in regulating DNA methylation, histone deacetylation, and mitochondrial biogenesis. These natural agents offer dual-action therapies by reducing oxidative stress and inflammation while promoting neuronal survival and remyelination. This review explores the therapeutic potential of herbal drugs targeting epigenetic and mitochondrial pathways in MS, evaluating their mechanisms of action and highlighting their promise as novel therapeutic agents. While initial findings are encouraging, further research and clinical trials are required to validate the efficacy of these herbal treatments and fully understand their potential in slowing disease progression and improving patient outcomes in MS. Such exploration could pave the way for safer, multi-targeted therapies, offering new hope in the management of MS and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ashwani
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | | | - Mayank Kumar Choudhary
- Department of Pharmacology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Dalapathi Gugulothu
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India.
| | - Deepti Pandita
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Surajpal Verma
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Lalitkumar K Vora
- School of Pharmacy, Medical Biology Centre, Queen'S University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK.
| | - Dharmendra Kumar Khatri
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India.
| | - Debapriya Garabadu
- Department of Pharmacology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
18
|
Hu F, Tong S, Xu H. Schisandrin B Improves Mitochondrial Function and Inhibits HT22 Cell Apoptosis by Regulating Sirt3 Protein. J Membr Biol 2025; 258:123-133. [PMID: 39939534 DOI: 10.1007/s00232-025-00340-9] [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: 12/13/2024] [Accepted: 01/25/2025] [Indexed: 02/14/2025]
Abstract
Neurological diseases refer to pathological changes that occur in the brain, spinal cord, and peripheral nerves. Their etiologies are complex, treatment outcomes are poor, and prognoses are unfavorable. Therefore, how to improve the treatment efficacy of neurological diseases is an urgent problem to be addressed in current clinical practice. Schisandrin B, a commonly used traditional Chinese medicine in clinical settings, has anti-tumor, anti-inflammatory, and wound-healing promoting effects. However, there are relatively few studies on its application in the treatment of neurological diseases. In this study, HT22 nerve cells were cultured, and an injury model was constructed by applying H2O2 stimulation to explore the protective effect of Schisandrin B on these cells. The research results showed that compared with the H2O2 group, Schisandrin B could significantly increase the viability (30.872%) and migration ability (42.756%) of HT22 cells, and inhibit the apoptosis of HT22 cells (22.817%). Further exploration of the mechanism revealed that Schisandrin B regulated the mitochondrial dynamic balance and membrane potential level of HT22 cells by upregulating the expression of Sirt3 protein, enhanced the mitochondrial energy metabolism (with an increase of 53.411% in ATP production), and maintained the integrity of the quantity and structure of mitochondria, ultimately exerting a protective effect on HT22 cells.
Collapse
Affiliation(s)
- Fei Hu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, Ningbo, China
| | - Songlin Tong
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, No. 999, South Second Ring Road, Hushan Street, Cixi, Ningbo, China
| | - Hongming Xu
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, No. 999, South Second Ring Road, Hushan Street, Cixi, Ningbo, China.
| |
Collapse
|
19
|
Dorémus L, Dugast E, Delafenêtre A, Delouche M, Aupy T, Bernard O, Sebille S, Thiriet N, Piquereau J. Optimization of permeabilized brain tissue preparation to improve the analysis of mitochondrial oxidative capacities in specific subregions of the rat brain. J Neurosci Methods 2025; 416:110387. [PMID: 39921038 DOI: 10.1016/j.jneumeth.2025.110387] [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: 12/18/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND As the major energy producer of cerebral tissue, mitochondria play key roles in brain physiology and physiopathology. Yet, the fine details of the functioning of mitochondrial oxidative phosphorylation in this organ are still scattered with grey area. This is partly due to the heterogeneity of this tissue that challenges our abilities to study specific cerebral subregions. In the last decades, cerebral mitochondria have largely been studied as a single entity by isolating mitochondria from large sections of brain. Given the evidence that these organelles must adapt to brain areas functions, it seems crucial to develop technologies enabling study of the mitochondria in given subregions. NEW METHOD A few years ago, a method allowing the investigation of mitochondrial functions in permeabilized brain subregions have been proposed by Holloway's team. Although this protocol represented a significant advance, we propose improvements in the tissue permeabilization procedure and in the conditions for measuring oxidative capacity. RESULTS AND COMPARISON WITH EXISTING METHODS The present study demonstrates that adjustments enabled obtention of higher respiration values than Holloway's protocol and might allow the detection of slight mitochondrial alterations. In a second part of this study, we showed that cortex, striatum, hippocampus and cerebellum displayed similar maximal oxidative capacities (under pyruvate, malate and succinate) while complex IV-driven respiration is significantly lower in cerebellum compared to cortex. These observations were supported by the measurement of citrate synthase and cytochrome oxidase activities. CONCLUSION The developed procedure improves the investigations of mitochondrial electron transfer chain in specific cerebral regions.
Collapse
Affiliation(s)
- Léa Dorémus
- Laboratoire PRéTI UR 24184, Université de Poitiers, Poitiers, France.
| | - Emilie Dugast
- INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Université de Poitiers, Poitiers, France.
| | | | - Morgane Delouche
- UMR-S 1180, Inserm, Univ. Paris-Sud, Université Paris-Saclay, Orsay, France.
| | - Thomas Aupy
- INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Université de Poitiers, Poitiers, France.
| | - Olivier Bernard
- Laboratoire Mobilité Vieillissement et Exercice, Université de Poitiers, Poitiers, France.
| | - Stéphane Sebille
- Laboratoire PRéTI UR 24184, Université de Poitiers, Poitiers, France.
| | - Nathalie Thiriet
- INSERM, U-1084, Laboratoire des Neurosciences Expérimentales et Cliniques, Université de Poitiers, Poitiers, France.
| | - Jérôme Piquereau
- Laboratoire PRéTI UR 24184, Université de Poitiers, Poitiers, France; UMR-S 1180, Inserm, Univ. Paris-Sud, Université Paris-Saclay, Orsay, France.
| |
Collapse
|
20
|
Wadan AHS, Shaaban AH, El-Sadek MZ, Mostafa SA, Moshref AS, El-Hussein A, Ellakwa DES, Mehanny SS. Mitochondrial-based therapies for neurodegenerative diseases: a review of the current literature. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04014-0. [PMID: 40163151 DOI: 10.1007/s00210-025-04014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Neurodegenerative disorders present significant challenges to modern medicine because of their complex etiology, pathogenesis, and progressive nature, which complicate practical treatment approaches. Mitochondrial dysfunction is an important contributor to the pathophysiology of various neurodegenerative illnesses, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). This review paper examines the current literature highlighting the multifaceted functions of mitochondria, including energy production, calcium signaling, apoptosis regulation, mitochondrial biogenesis, mitochondrial dynamics, axonal transport, endoplasmic reticulum-mitochondrial interactions, mitophagy, mitochondrial proteostasis, and their crucial involvement in neuronal health. The literature emphasizes the increasing recognition of mitochondrial dysfunction as a critical factor in the progression of neurodegenerative disorders, marking a shift from traditional symptom management to innovative mitochondrial-based therapies. By discussing mitochondrial mechanisms, including mitochondrial quality control (MQC) processes and the impact of oxidative stress, this review highlights the need for novel therapeutic strategies to restore mitochondrial function, protect neuronal connections and integrity, and slow disease progression. This comprehensive review aims to provide insights into potential interventions that could transform the treatment landscape for neurodegenerative diseases, addressing symptoms and underlying pathophysiological changes.
Collapse
Affiliation(s)
- Al-Hassan Soliman Wadan
- Department of Oral Biology, Faculty of Dentistry, Galala University, Galala Plateau, Attaka, Suez Governorate, 15888, Egypt.
| | - Ahmed H Shaaban
- Department of Biology, Faculty of Science, Galala University, Galala Plateau, Attaka,, Suez Governorate, 15888, Egypt
| | - Mohamed Z El-Sadek
- Department of Biology, Faculty of Science, Galala University, Galala Plateau, Attaka,, Suez Governorate, 15888, Egypt
| | | | - Ahmed Sherief Moshref
- Faculty of Dentistry, Galala University, Galala Plateau, Attaka, Suez Governorate, 15888, Egypt
| | - Ahmed El-Hussein
- Department of Biology, Faculty of Science, Galala University, Galala Plateau, Attaka,, Suez Governorate, 15888, Egypt
- Department of Laser Applications in Meteorology, Photochemistry, and Biotechnology, The National Institute of Laser Enhanced Science, Cairo University, Cairo, 11316, Egypt
| | - Doha El-Sayed Ellakwa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt
| | - Samah S Mehanny
- Department of Oral Biology, Faculty of Dentistry, Galala University, Galala Plateau, Attaka, Suez Governorate, 15888, Egypt
- Department of Oral Biology, Faculty of Dentistry, Cairo University, Cairo, Egypt
| |
Collapse
|
21
|
Nibrad D, Shiwal A, Tadas M, Katariya R, Kale M, Kotagale N, Umekar M, Taksande B. Therapeutic modulation of mitochondrial dynamics by agmatine in neurodegenerative disorders. Neuroscience 2025; 569:43-57. [PMID: 39890051 DOI: 10.1016/j.neuroscience.2025.01.061] [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: 12/10/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Mitochondrial dysfunction is a pivotal factor in the pathogenesis of neurodegenerative disorders, driving neuronal degeneration through mechanisms involving oxidative stress, impaired energy production, and dysregulated calcium homeostasis. Agmatine, an endogenous polyamine derived from arginine, has garnered attention for its neuroprotective properties, including anti-inflammatory, anti-oxidative, and antiapoptotic effects. Recent studies have highlighted the potential of agmatine in preserving mitochondrial function and mitigating neurodegeneration, making it a promising candidate for therapeutic intervention. One of the key mechanisms by which agmatine exerts its neuroprotective effects is through the maintenance of mitochondrial homeostasis. Agmatine has been shown to modulate mitochondrial dynamics, promoting mitochondrial fusion and fission balance essential for cellular energy metabolism and signaling. Moreover, agmatine acts as a regulator of mitochondrial permeability transition pore (mPTP) opening, preventing excessive calcium influx and subsequent mitochondrial dysfunction. Despite promising findings, challenges such as optimizing agmatine's pharmacokinetics, determining optimal dosing regimens, and elucidating its precise molecular targets within mitochondria remain to be addressed. Future research directions should focus on developing targeted delivery systems for agmatine, investigating its interactions with mitochondrial proteins, and conducting well-designed clinical trials to evaluate its therapeutic efficacy and safety profile in neurodegenerative disorders. Overall, agmatine emerges as a novel therapeutic agent with the potential to modulate mitochondrial homeostasis and alleviate neurodegenerative pathology, offering new avenues for treating these debilitating conditions.
Collapse
Affiliation(s)
- Dhanshree Nibrad
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Amit Shiwal
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Manasi Tadas
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Raj Katariya
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Mayur Kale
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy, Kathora Naka, VMV Road, Amravati, (M.S.) 444604, India
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India.
| |
Collapse
|
22
|
Mozafari S, Peruzzotti-Jametti L, Pluchino S. Mitochondria transfer for myelin repair. J Cereb Blood Flow Metab 2025:271678X251325805. [PMID: 40079508 PMCID: PMC11907575 DOI: 10.1177/0271678x251325805] [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: 01/06/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/15/2025]
Abstract
Demyelination is a common feature of neuroinflammatory and degenerative diseases of the central nervous system (CNS), such as multiple sclerosis (MS). It is often linked to disruptions in intercellular communication, bioenergetics and metabolic balance accompanied by mitochondrial dysfunction in cells such as oligodendrocytes, neurons, astrocytes, and microglia. Although current MS treatments focus on immunomodulation, they fail to stop or reverse demyelination's progression. Recent advancements highlight intercellular mitochondrial exchange as a promising therapeutic target, with potential to restore metabolic homeostasis, enhance immunomodulation, and promote myelin repair. With this review we will provide insights into the CNS intercellular metabolic decoupling, focusing on the role of mitochondrial dysfunction in neuroinflammatory demyelinating conditions. We will then discuss emerging cell-free biotherapies exploring the therapeutic potential of transferring mitochondria via biogenic carriers like extracellular vesicles (EVs) or synthetic liposomes, aimed at enhancing mitochondrial function and metabolic support for CNS and myelin repair. Lastly, we address the key challenges for the clinical application of these strategies and discuss future directions to optimize mitochondrial biotherapies. The advancements in this field hold promise for restoring metabolic homeostasis, and enhancing myelin repair, potentially transforming the therapeutic landscape for neuroinflammatory and demyelinating diseases.
Collapse
Affiliation(s)
- Sabah Mozafari
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
Nishikiori N, Watanabe M, Higashide M, Umetsu A, Ogawa T, Furuhashi M, Ohguro H, Sato T. The Combination of PPARα Agonist GW7647 and Imeglimin Has Potent Effects on High-Glucose-Induced Cellular Biological Responses in Human Retinal Pigment Epithelium Cells. Bioengineering (Basel) 2025; 12:265. [PMID: 40150729 PMCID: PMC11939608 DOI: 10.3390/bioengineering12030265] [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: 11/01/2024] [Revised: 01/12/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Hyperglycemic changes in the cellular biological properties of retinal pigment epithelium cells are involved in the pathophysiology of diabetic retinopathy (DR). To assess the effects of the new anti-diabetic agent imeglimin (Ime) on DR, the pharmacological effects of Ime and those of metformin (Met) in combination with the PPARα agonist GW7646 (GW) on adult retinal pigment epithelium (ARPE19) cells cultured in high-glucose conditions were compared. METHODS Cell viability, levels of reactive oxygen species (ROS), monolayer barrier function measured by transepit very much helial electrical resistance (TEER), and metabolic functions determined by an extracellular flux analyzer were evaluated. RESULTS While glucose concentrations did not alter cell viability regardless of the presence of Met or Ime, levels of ROS were significantly increased by the high-glucose conditions, and increased levels of ROS were significantly alleviated by the combination of Ime and GW but not by Met alone. Similarly, TEER values were increased by high-glucose conditions, but the effects of high-glucose conditions were dramatically enhanced by the combination of Ime and GW. Furthermore, a metabolic assay showed that an energetic shift was induced by the combination of Ime and GW, whereas energy status became quiescent with Met or Ime alone. CONCLUSIONS The collective results suggest that Ime in combination with GW has synergetic effects on high-glucose-induced cellular biological changes in ARPE19 cells.
Collapse
Affiliation(s)
- Nami Nishikiori
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.); (A.U.)
| | - Megumi Watanabe
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.); (A.U.)
| | - Megumi Higashide
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.); (A.U.)
| | - Araya Umetsu
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.); (A.U.)
| | - Toshifumi Ogawa
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.O.); (M.F.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.W.); (M.H.); (A.U.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| |
Collapse
|
24
|
Chesebro AG, Antal BB, Weistuch C, Mujica-Parodi LR. Challenges and Frontiers in Computational Metabolic Psychiatry. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2025; 10:258-266. [PMID: 39481469 DOI: 10.1016/j.bpsc.2024.10.011] [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: 08/08/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
One of the primary challenges in metabolic psychiatry is that the disrupted brain functions that underlie psychiatric conditions arise from a complex set of downstream and feedback processes that span multiple spatiotemporal scales. Importantly, the same circuit can have multiple points of failure, each of which results in a different type of dysregulation, and thus elicits distinct cascades downstream that produce divergent signs and symptoms. Here, we illustrate this challenge by examining how subtle differences in circuit perturbations can lead to divergent clinical outcomes. We also discuss how computational models can perform the spatially heterogeneous integration and bridge in vitro and in vivo paradigms. By leveraging recent methodological advances and tools, computational models can integrate relevant processes across scales (e.g., tricarboxylic acid cycle, ion channel, neural microassembly, whole-brain macrocircuit) and across physiological systems (e.g., neural, endocrine, immune, vascular), providing a framework that can unite these mechanistic processes in a manner that goes beyond the conceptual and descriptive to the quantitative and generative. These hold the potential to sharpen our intuitions toward circuit-based models for personalized diagnostics and treatment.
Collapse
Affiliation(s)
- Anthony G Chesebro
- Department of Biomedical Engineering and Laufer Center for Physical and Quantitative Biology, Renaissance School of Medicine, State University of New York at Stony Brook, Stony Brook, New York; Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Botond B Antal
- Department of Biomedical Engineering and Laufer Center for Physical and Quantitative Biology, Renaissance School of Medicine, State University of New York at Stony Brook, Stony Brook, New York; Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Corey Weistuch
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lilianne R Mujica-Parodi
- Department of Biomedical Engineering and Laufer Center for Physical and Quantitative Biology, Renaissance School of Medicine, State University of New York at Stony Brook, Stony Brook, New York; Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts; Santa Fe Institute, Santa Fe, New Mexico.
| |
Collapse
|
25
|
Campomayor NB, Kim HJ, Kim M. Pro-Oxidative and Inflammatory Actions of Extracellular Hemoglobin and Heme: Molecular Events and Implications for Alzheimer's and Parkinson Disease. Biomol Ther (Seoul) 2025; 33:235-248. [PMID: 39962769 PMCID: PMC11893490 DOI: 10.4062/biomolther.2024.224] [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: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 03/01/2025] Open
Abstract
Hemoglobin (Hb) and heme, which are typically confined within red blood cells (RBCs), are essential for intravascular transport of gases and nutrients. However, these molecules acquire secondary functions upon exposure to the extracellular environment. Hb and heme generate reactive oxygen species (ROS), which are potent pro-inflammatory agents that contribute to oxidative stress and cellular damage. These events are relevant to neurodegenerative processes, where oxidative stress, irregular deposition of protein aggregates, and chronic inflammation are key pathological features. Extracellular Hb, heme, and oxidative stress derived from hemorrhagic events or RBC lysis may contribute to increased blood-brain barrier (BBB) permeability. These events allow Hb and heme to interact with neuroimmune cells and pathological protein aggregates, further amplifying pro-inflammatory signaling and the progression of Alzheimer's disease (AD) and Parkinson disease (PD). Chronic neuroinflammation, oxidative stress, and mitochondrial dysfunction lead to neuronal degeneration. Here, we sought to elucidate the pro-oxidative and inflammatory actions of extracellular Hb and heme, emphasizing their potential impact on AD and PD development.
Collapse
Affiliation(s)
- Nicole Bon Campomayor
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Chemistry & Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Chemistry & Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
26
|
Jirström E, Matveeva A, Baindoor S, Donovan P, Ma Q, Morrissey EP, Arijs I, Boeckx B, Lambrechts D, Garcia-Munoz A, Dillon ET, Wynne K, Ying Z, Matallanas D, Hogg MC, Prehn JHM. Effects of ALS-associated 5'tiRNA Gly-GCC on the transcriptomic and proteomic profile of primary neurons in vitro. Exp Neurol 2025; 385:115128. [PMID: 39719207 DOI: 10.1016/j.expneurol.2024.115128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
tRNA-derived stress-induced RNAs (tiRNAs) are a new class of small non-coding RNA that have emerged as important regulators of cellular stress responses. tiRNAs are derived from specific tRNA cleavage by the stress-induced ribonuclease angiogenin (ANG). Loss-of-function mutations in the ANG gene are linked to amyotrophic lateral sclerosis (ALS), and elevated levels of specific tiRNAs were recently identified in ALS patient serum samples. However, the biological role of tiRNA production in neuronal stress responses and neurodegeneration remains largely unknown. Here, we investigated the genome-wide regulation of neuronal stress responses by a specific tiRNA, 5'tiRNAGly-GCC, which we found to be upregulated in primary neurons exposed to ALS-relevant stresses and in the spinal cord of three ALS mouse models. Whole-transcript RNA sequencing and label-free mass spectrometry on primary neurons transfected with a synthetic mimic of 5'tiRNAGly-GCC revealed predominantly downregulated RNA and protein levels, with more pronounced changes in the proteome. Over half of the downregulated mRNAs contained predicted 5'tiRNAGly-GCC binding sites, indicating that this tiRNA may silence target genes via complementary binding. On the proteome level, we observed reduction in proteins involved in translation initiation and ribosome assembly, pointing to inhibitory effects on translation. Together, these findings suggest that 5'tiRNAGly-GCC is an ALS-associated tiRNA that functions to fine-tune gene expression and supress protein synthesis as part of an ANG-induced neuronal stress response.
Collapse
Affiliation(s)
- Elisabeth Jirström
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Anna Matveeva
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Sharada Baindoor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Paul Donovan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Qilian Ma
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Elena Perez Morrissey
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Ingrid Arijs
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology, Leuven, Belgium
| | - Bram Boeckx
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Center for Cancer Biology, Leuven, Belgium
| | - Amaya Garcia-Munoz
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eugène T Dillon
- Mass Spectrometry Resource, Conway Institute of Biomolecular & Biomedical Research, University College Dublin 4, Ireland
| | - Kieran Wynne
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Zheng Ying
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Marion C Hogg
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland; FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| |
Collapse
|
27
|
Sil R, Chakraborti AS. Major heme proteins hemoglobin and myoglobin with respect to their roles in oxidative stress - a brief review. Front Chem 2025; 13:1543455. [PMID: 40070406 PMCID: PMC11893434 DOI: 10.3389/fchem.2025.1543455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Oxidative stress is considered as the root-cause of different pathological conditions. Transition metals, because of their redox-active states, are capable of free radical generation contributing oxidative stress. Hemoglobin and myoglobin are two major heme proteins, involved in oxygen transport and oxygen storage, respectively. Heme prosthetic group of heme proteins is a good reservoir of iron, the most abundant transition metal in human body. Although iron is tightly bound in the heme pocket of these proteins, it is liberated under specific circumstances yielding free ferrous iron. This active iron can react with H2O2, a secondary metabolite, forming hydroxyl radical via Fenton reaction. Hydroxyl radical is the most harmful free radical among all the reactive oxygen species. It causes oxidative stress by damaging lipid membranes, proteins and nucleic acids, activating inflammatory pathways and altering membrane channels, resulting disease conditions. In this review, we have discussed how heme-irons of hemoglobin and myoglobin can promote oxidative stress under different pathophysiological conditions including metabolic syndrome, diabetes, cardiovascular, neurodegenerative and renal diseases. Understanding the association of heme proteins to oxidative stress may be important for knowing the complications as well as therapeutic management of different pathological conditions.
Collapse
Affiliation(s)
| | - Abhay Sankar Chakraborti
- Department of Biophysics, Molecular Biology and Bioinformatics, University College of Science, University of Calcutta, Kolkata, India
| |
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
|
Lin S, Hu G, Zhang M, Li J. ATP Binding and Inhibition of Intrinsically Disordered Protein Interactions. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:3315-3324. [PMID: 39885825 DOI: 10.1021/acs.langmuir.4c04216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Recent studies have shown that ATP at high physiological concentrations (>5 mM) can inhibit liquid-liquid phase separation (LLPS) driven by interactions between intrinsically disordered proteins (IDPs). However, the mechanism underlying such inhibitory effect still remains elusive. Here, we used all-atom molecular dynamics simulation to study the interaction of ATP with two typical IDPs (i.e., FUS PLD and RGG domain of hnRNP G), and its impacts on IDP interactions. ATP exhibits a considerable tendency to bind to both IDPs and effectively inhibits their interactions. For the RGG domain, Arg residues are critical for both ATP binding and IDP interactions. The inhibitory effect of ATP is largely attributed to its competitive binding mode to Arg residues. Similar competitive binding of ATP is also observed in FUS PLD. Both ATP binding and the PLD interaction share the residues including Gln, Ser, and Tyr residues, while the competition is rather modest due to the abundance of these residues in the sequence. Interestingly, ATP undergoes considerable diffusion on the surface of PLD, which is an order of magnitude faster than the evolution of the contact area of PLDs. The temporal separation of these two processes remarkably promotes the inhibitory effect of ATP on PLD interaction. Given the representativeness of these two IDPs, competitive binding may serve as a general mechanism underlying ATP inhibition on IDP interactions at high physiological levels.
Collapse
Affiliation(s)
- Shiyan Lin
- Zhejiang Province Key Laboratory of Quantum Technology and Device, School of Physics, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Guorong Hu
- Zhejiang Province Key Laboratory of Quantum Technology and Device, School of Physics, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Moxin Zhang
- Zhejiang Province Key Laboratory of Quantum Technology and Device, School of Physics, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Jingyuan Li
- Zhejiang Province Key Laboratory of Quantum Technology and Device, School of Physics, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| |
Collapse
|
30
|
Gil J, Kim D, Choi S, Bae ON. Cadmium-induced iron dysregulation contributes to functional impairment in brain endothelial cells via the ferroptosis pathway. Toxicol Appl Pharmacol 2025; 495:117233. [PMID: 39842614 DOI: 10.1016/j.taap.2025.117233] [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: 10/04/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Cadmium (Cd2+) is a heavy metal that is a major hazardous environmental contaminant, ubiquitously present in the environment. Cd2+ exposure has been closely associated with an increased prevalence and severity of neurological and cardiovascular diseases (CVD). The blood-brain barrier (BBB) plays a crucial role in protecting the brain from external environmental factors. Mitochondria play an important role in maintaining the barrier function of brain endothelial cells by regulating energy metabolism and redox homeostasis. In this study, we aimed to assess the cytotoxic effects of Cd2+ on the integrity and function of brain endothelial cells. After 24 h of exposure, Cd2+ reduced cell survival, tight junction protein expression, and trans-endothelial electrical resistance (TEER) in bEnd.3 cells suggest a potential BBB integrity disruption by Cd2+ exposure. To clarify the underlying mechanism, we further investigated the role of mitochondria in iron overload-mediated cell death following Cd2+ exposure. Cd2+ induced a substantial reduction in mitochondrial basal respiration and ATP production in brain endothelial cells, suggesting mitochondrial dysfunction. In addition, Cd2+ exposure led to impaired autophagy, elevated iron levels, and increased lipid peroxidation, indicating the initiation of ferroptosis, a form of cell death triggered by iron. In summary, our research suggests that Cd2+ exposure can disrupt BBB function by causing mitochondrial dysfunction and disrupting iron homeostasis.
Collapse
Affiliation(s)
- Junkyung Gil
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Donghyun Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Sungbin Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| |
Collapse
|
31
|
Mohammad ZB, Yudin SCY, Goldberg BJ, Serra KL, Klegeris A. Exploring neuroglial signaling: diversity of molecules implicated in microglia-to-astrocyte neuroimmune communication. Rev Neurosci 2025; 36:91-117. [PMID: 39240134 PMCID: PMC11717358 DOI: 10.1515/revneuro-2024-0081] [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: 06/10/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024]
Abstract
Effective communication between different cell types is essential for brain health, and dysregulation of this process leads to neuropathologies. Brain glial cells, including microglia and astrocytes, orchestrate immune defense and neuroimmune responses under pathological conditions during which interglial communication is indispensable. Our appreciation of the complexity of these processes is rapidly increasing due to recent advances in molecular biology techniques, which have identified numerous phenotypic states of both microglia and astrocytes. This review focuses on microglia-to-astrocyte communication facilitated by secreted neuroimmune modulators. The combinations of interleukin (IL)-1α, tumor necrosis factor (TNF), plus complement component C1q as well as IL-1β plus TNF are already well-established microglia-derived stimuli that induce reactive phenotypes in astrocytes. However, given the large number of inflammatory mediators secreted by microglia and the rapidly increasing number of distinct functional states recognized in astrocytes, it can be hypothesized that many more intercellular signaling molecules exist. This review identifies the following group of cytokines and gliotransmitters that, while not established as interglial mediators yet, are known to be released by microglia and elicit functional responses in astrocytes: IL-10, IL-12, IL-18, transforming growth factor (TGF)-β, interferon (IFN)-γ, C-C motif chemokine ligand (CCL)5, adenosine triphosphate (ATP), l-glutamate, and prostaglandin E2 (PGE2). The review of molecular mechanisms engaged by these mediators reveals complex, partially overlapping signaling pathways implicated in numerous neuropathologies. Additionally, lack of human-specific studies is identified as a significant knowledge gap. Further research on microglia-to-astrocyte communication is warranted, as it could discover novel interglial signaling-targeted therapies for diverse neurological disorders.
Collapse
Affiliation(s)
- Zainab B. Mohammad
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Samantha C. Y. Yudin
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Benjamin J. Goldberg
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Kursti L. Serra
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Andis Klegeris
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| |
Collapse
|
32
|
Nasme F, Behera J, Tyagi P, Debnath N, Falcone JC, Tyagi N. The potential link between the development of Alzheimer's disease and osteoporosis. Biogerontology 2025; 26:43. [PMID: 39832071 DOI: 10.1007/s10522-024-10181-z] [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: 10/10/2024] [Accepted: 12/28/2024] [Indexed: 01/22/2025]
Abstract
Alzheimer's disease (AD) and osteoporosis (OP) pose distinct but interconnected health challenges, both significantly impacting the aging population. AD, a neurodegenerative disorder characterized by memory impairment and cognitive decline, is primarily associated with the accumulation of abnormally folded amyloid beta (Aβ) peptides and neurofibrillary tangles in the brain. OP, a skeletal disorder marked by low bone mineral density, involves dysregulation of bone remodeling and is associated with an increased risk of fractures. Recent studies have revealed an intriguing link between AD and OP, highlighting shared pathological features indicative of common regulatory pathophysiological pathways. In this article, we elucidate the signaling mechanisms that regulate the pathology of AD and OP and offer insights into the intricate network of factors contributing to these conditions. We also examine the role of bone-derived factors in the progression of AD, underscoring the plausibility of bidirectional communication between the brain and the skeletal system. The presence of amyloid plaques in the brain of individuals with AD is akin to the accumulation of brain Aβ in vascular dementia, pointing towards the need for further investigation of shared molecular mechanisms. Moreover, we discuss the role of bone-derived microRNAs that may regulate the pathological progression of AD, providing a novel perspective on the role of skeletal factors in neurodegenerative diseases. The insights presented here should help researchers engaged in exploring innovative therapeutic approaches targeting both neurodegenerative and skeletal disorders in aging populations.
Collapse
Affiliation(s)
- Fariha Nasme
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Jyotirmaya Behera
- Division of Immunology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Prisha Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Rahya-Suchani (Bagla) Samba, Jammu, Jammu & Kashmir, 181143, India
| | - Jeff C Falcone
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Neetu Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
33
|
Kim S, Jung UJ, Kim SR. The Crucial Role of the Blood-Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications. J Clin Med 2025; 14:386. [PMID: 39860392 PMCID: PMC11765772 DOI: 10.3390/jcm14020386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The blood-brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer's and Parkinson's disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
34
|
Vega-Letter AM, García-Guerrero C, Yantén-Fuentes L, Pradenas C, Herrera-Luna Y, Lara-Barba E, Bustamante-Barrientos FA, Rojas M, Araya MJ, Jeraldo N, Aros C, Troncoso F, Poblete D, Court A, Ortloff A, Barraza J, Velarde F, Farkas C, Carril C, Luque-Campos N, Almarza G, Barahona M, Matas J, Cereceda L, Lorca R, Toledo J, Oyarce K, Vernal R, Caicedo A, Del Campo A, Hidalgo Y, Elizondo-Vega R, Djouad F, Khoury M, Figueroa FE, Luz-Crawford P. Safety and efficacy of mesenchymal stromal cells mitochondria transplantation as a cell-free therapy for osteoarthritis. J Transl Med 2025; 23:26. [PMID: 39773289 PMCID: PMC11706173 DOI: 10.1186/s12967-024-05945-7] [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: 06/10/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE The inflammatory responses from synovial fibroblasts and macrophages and the mitochondrial dysfunction in chondrocytes lead to oxidative stress, disrupt extracellular matrix (ECM) homeostasis, and accelerate the deterioration process of articular cartilage in osteoarthritis (OA). In recent years, it has been proposed that mesenchymal stromal cells (MSC) transfer their functional mitochondria to damaged cells in response to cellular stress, becoming one of the mechanisms underpinning their therapeutic effects. Therefore, we hypothesize that a novel cell-free treatment for OA could involve direct mitochondria transplantation, restoring both cellular and mitochondrial homeostasis. METHODS Mitochondria were isolated from Umbilical Cord (UC)-MSC (Mito-MSC) and characterized based on their morphology, phenotype, functions, and their ability to be internalized by different articular cells. Furthermore, the transcriptional changes following mitochondrial uptake by chondrocytes were evaluated using an Affymetrix analysis, Lastly, the dose dependence therapeutic efficacy, biodistribution and immunogenicity of Mito-MSC were assessed in vivo, through an intra-articular injection in male C57BL6 mice in a collagenase-induced OA (CIOA) model. RESULTS Our findings demonstrate the functional integrity of Mito-MSC and their ability to be efficiently transferred into chondrocytes, synovial macrophages, and synovial fibroblasts. Moreover, the transcriptomic analysis showed the upregulation of genes involved in stress such as DNA reparative machinery and inflammatory antiviral responses. Finally, Mito-MSC transplantation yielded significant reductions in joint mineralization, a hallmark of OA progression, as well as improvements in OA-related histological signs, with the lower dose exhibiting better therapeutic efficacy. Furthermore, Mito-MSC was detected within the knee joint for up to 24 h post-injection without eliciting an inflammatory response in CIOA mice. CONCLUSION Collectively, our results reveal that mitochondria derived from MSC are transferred to key articular cells and are retained in the joint without generating an inflammatory immune response mitigating articular cartilage degradation in OA, probably through a restorative effect triggered by the stress antiviral response within OA chondrocytes.
Collapse
Affiliation(s)
- Ana Maria Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Cynthia García-Guerrero
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Liliana Yantén-Fuentes
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Carolina Pradenas
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Yeimi Herrera-Luna
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Eliana Lara-Barba
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Masyelly Rojas
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - María Jesús Araya
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Nicole Jeraldo
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Red de Equipamiento Científico Avanzado-REDECA, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Constanza Aros
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Francisca Troncoso
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Daniela Poblete
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Angela Court
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Jose Barraza
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Francesca Velarde
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Carlos Farkas
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Claudio Carril
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Chile
| | - Noymar Luque-Campos
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Gonzalo Almarza
- Laboratorio de Fisiología y Bioenergetica Celular, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maximiliano Barahona
- Departamento de Ortopedia y Traumatología, Hospital Clinico Universidad de Chile, Independencia, Chile
| | - Jose Matas
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
| | - Lucas Cereceda
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Rocío Lorca
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado-REDECA, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Karina Oyarce
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Chile
| | - Rolando Vernal
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Andrés Caicedo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud e Instituto de Investigaciones en Biomedicina iBioMed, Escuela de Medicina, Quito, Ecuador-Mito-Act Research Consortium, Quito, Ecuador
| | - Andrea Del Campo
- Laboratorio de Fisiología y Bioenergetica Celular, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yessia Hidalgo
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Farida Djouad
- IRMB, Université de Montpellier, INSERM, Montpellier, France
- CHU Montpellier, Montpellier, France
| | - Maroun Khoury
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile.
| | - Fernando E Figueroa
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile.
| | - Patricia Luz-Crawford
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
| |
Collapse
|
35
|
Ferretti S, Zanella I. The Underestimated Role of Iron in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:12987. [PMID: 39684697 DOI: 10.3390/ijms252312987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The term frontotemporal dementia (FTD) comprises a group of neurodegenerative disorders characterized by the progressive degeneration of the frontal and temporal lobes of the brain with language impairment and changes in cognitive, behavioral and executive functions, and in some cases motor manifestations. A high proportion of FTD cases are due to genetic mutations and inherited in an autosomal-dominant manner with variable penetrance depending on the implicated gene. Iron is a crucial microelement that is involved in several cellular essential functions in the whole body and plays additional specialized roles in the central nervous system (CNS) mainly through its redox-cycling properties. Such a feature may be harmful under aerobic conditions, since it may lead to the generation of highly reactive hydroxyl radicals. Dysfunctions of iron homeostasis in the CNS are indeed involved in several neurodegenerative disorders, although it is still challenging to determine whether the dyshomeostasis of this essential but harmful metal is a direct cause of neurodegeneration, a contributor factor or simply a consequence of other neurodegenerative mechanisms. Unlike many other neurodegenerative disorders, evidence of the dysfunction in brain iron homeostasis in FTD is still scarce; nonetheless, the recent literature intriguingly suggests its possible involvement. The present review aims to summarize what is currently known about the contribution of iron dyshomeostasis in FTD based on clinical, imaging, histological, biochemical and molecular studies, further suggesting new perspectives and offering new insights for future investigations on this underexplored field of research.
Collapse
Affiliation(s)
- Sara Ferretti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Medical Genetics Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
36
|
Bernardo G, Prado MA, Dashtmian AR, Favaro M, Mauri S, Borsetto A, Marchesan E, Paulo JA, Gygi SP, Finley DJ, Ziviani E. USP14 inhibition enhances Parkin-independent mitophagy in iNeurons. Pharmacol Res 2024; 210:107484. [PMID: 39486496 DOI: 10.1016/j.phrs.2024.107484] [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: 07/31/2024] [Revised: 10/20/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Loss of proteostasis is well documented during physiological aging and depends on the progressive decline in the activity of two major degradative mechanisms: the ubiquitin-proteasome system (UPS) and the autophagy-lysosomal pathway. This decline in proteostasis is exacerbated in age-associated neurodegenerative diseases, such as Parkinson's Disease (PD). In PD, patients develop an accumulation of aggregated proteins and dysfunctional mitochondria, which leads to ROS production, neuroinflammation and neurodegeneration. We recently reported that inhibition of the deubiquitinating enzyme USP14, which is known to enhance both the UPS and autophagy, increases lifespan and rescues the pathological phenotype of two Drosophila models of PD. Studies on the effects of USP14 inhibition in mammalian neurons have not yet been conducted. To close this gap, we exploited iNeurons differentiated from human embryonic stem cells (hESCs), and investigated the effect of inhibiting USP14 in these cultured neurons. Quantitative global proteomics analysis performed following genetic ablation or pharmacological inhibition of USP14 demonstrated that USP14 loss of function specifically promotes mitochondrial autophagy in iNeurons. Biochemical and imaging data also showed that USP14 inhibition enhances mitophagy. The mitophagic effect of USP14 inhibition proved to be PINK1/Parkin- independent, instead relying on expression of the mitochondrial E3 Ubiquitin Ligase MITOL/MARCH5. Notably, USP14 inhibition normalized the mitochondrial defects of Parkin KO human neurons.
Collapse
Affiliation(s)
- Greta Bernardo
- Department of Biology, University of Padova, Padova, Italy
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, USA; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | | | - Sofia Mauri
- Department of Biology, University of Padova, Padova, Italy
| | - Alice Borsetto
- Department of Biology, University of Padova, Padova, Italy
| | | | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, USA
| | - Steve P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, USA
| | - Daniel J Finley
- Department of Cell Biology, Harvard Medical School, Boston, USA
| | - Elena Ziviani
- Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
37
|
Seddon AR, MacArthur CP, Hampton MB, Stevens AJ. Inflammation and DNA methylation in Alzheimer's disease: mechanisms of epigenetic remodelling by immune cell oxidants in the ageing brain. Redox Rep 2024; 29:2428152. [PMID: 39579010 PMCID: PMC11587723 DOI: 10.1080/13510002.2024.2428152] [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: 11/24/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease involving memory impairment, confusion, and behavioural changes. The disease is characterised by the accumulation of amyloid beta plaques and neurofibrillary tangles in the brain, which disrupt normal neuronal function. There is no known cure for Alzheimer's disease and due to increasing life expectancy, occurrence is projected to rise over the coming decades. The causes of Alzheimer's disease are multifactorial with inflammation, oxidative stress, genetic and epigenetic variation, and cerebrovascular abnormalities among the strongest contributors. We review the current literature surrounding inflammation and epigenetics in Alzheimer's disease, with a focus on how oxidants from infiltrating immune cells have the potential to alter DNA methylation profiles in the ageing brain.
Collapse
Affiliation(s)
- A. R. Seddon
- Mātai Hāora – Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| | - C. P. MacArthur
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| | - M. B. Hampton
- Mātai Hāora – Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - A. J. Stevens
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| |
Collapse
|
38
|
Yan X, Bai X, Sun G, Duan Z, Fu R, Zeng W, Zhu C, Fan D. Ginsenoside compound K alleviates brain aging by inhibiting ferroptosis through modulation of the ASK1-MKK7-JNK signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156239. [PMID: 39547099 DOI: 10.1016/j.phymed.2024.156239] [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: 08/16/2024] [Revised: 10/26/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Aging of the brain is a major contributor to the onset and progression of neurodegenerative diseases. Conventional treatments for these diseases are often limited by significant side effects and a lack of efficacy in halting disease progression. Ginsenoside compound K (CK), a bioactive secondary metabolite derived from ginseng, has shown promise because of its potent antioxidant properties. PURPOSE This study aimed to elucidate the molecular mechanisms underlying the impact of CK on brain senescence, with a particular focus on its role in modulating oxidative stress and related signaling pathways. METHODS We employed a d-galactose (D-gal)-induced PC-12 senescent cell model and a mouse brain aging model to explore the antioxidant properties of CK in the context of brain aging. The effects of CK on mitochondrial dysfunction associated with brain aging were assessed using immunofluorescence and western blotting techniques. The potential molecular mechanisms by CK influences brain aging were investigated using transcriptomic analysis and western blotting. Additionally, CK's regulatory effect on apoptosis signal-regulating kinase 1 (ASK1) was validated by molecular docking, microscale thermophoresis, and small interfering RNA transfection. RESULTS Our findings demonstrate that CK effectively alleviates cognitive decline associated with brain aging. CK reduces the number of senescent cells, alleviates neuronal damage, and enhances the activity of key antioxidant enzymes, including catalase, superoxide dismutase, and glutathione peroxidase. Additionally, CK restores mitochondrial function and upregulated the expression of solute carrier family 7 member 11 and glutathione peroxidase 4, thereby inhibiting ferroptosis. Furthermore, CK targets ASK1 and suppresses the hyperphosphorylation of MAPK kinase 7 (MKK7) and c-Jun N-terminal kinase (JNK). This suppression promotes the nuclear accumulation of nuclear factor erythroid 2-related factor 2 (Nrf2), effectively reducing ferroptosis and oxidative damage linked to brain aging. CONCLUSION In summary, our research demonstrates that CK effectively delays brain aging by inhibiting the ASK1-MKK7-JNK signaling pathway, enhancing nuclear Nrf2 expression, and suppressing the ferroptosis response. These findings highlight CK as a promising therapeutic agent for slowing brain aging and alleviating neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaojun Yan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Xue Bai
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Guanghui Sun
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Rongzhan Fu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| | - Wen Zeng
- Honghui Hospital, Xi' an Jiaotong University 710054, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China.
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China.
| |
Collapse
|
39
|
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
|
40
|
Adlimoghaddam A, Fayazbakhsh F, Mohammadi M, Babaei Z, Behrooz AB, Tabasi F, Guan T, Beheshti I, Aghaei M, Klionsky DJ, Albensi BC, Ghavami S. Sex and Region-Specific Disruption of Autophagy and Mitophagy in Alzheimer's Disease: Linking Cellular Dysfunction to Cognitive Decline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621097. [PMID: 39554142 PMCID: PMC11565785 DOI: 10.1101/2024.10.30.621097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Macroautophagy and mitophagy are critical processes in Alzheimer's disease (AD), yet their links to behavioral outcomes, particularly sex-specific differences, are not fully understood. This study investigates autophagy (LC3B-II, SQSTM1) and mitophagy (BNIP3L, BNIP3, BCL2L13) markers in the cortex and hippocampus of male and female 3xTg-AD mice, using western blotting, transmission electron microscopy (TEM), and behavioral tests (novel object recognition and novel object placement). Significant sex-specific differences emerged: female 3xTg-AD mice exhibited autophagosome accumulation due to impaired degradation in the cortex, while males showed fewer autophagosomes, especially in the hippocampus, without significant degradation changes. TEM analyses demonstrated variations in mitochondrial and mitophagosome numbers correlated with memory outcomes. Females had enhanced mitophagy, with higher BNIP3L and BCL2L13 levels, whereas males showed elevated BNIP3 dimers. Cognitive deficits in females correlated with mitochondrial dysfunction in the cortex, while in males, higher LC3B-II levels associated positively with cognitive performance, suggesting protective autophagy effects. Using machine learning, we predicted mitophagosome and mitochondrial numbers based on behavioral data, pioneering a predictive approach to cellular outcomes in AD. These findings underscore the importance of sex-specific regulation of autophagy and mitophagy in AD and support personalized therapeutic approaches targeting these pathways. Integrating machine learning emphasizes its potential to advance neurodegenerative research. Abstract Figure
Collapse
|
41
|
Stoccoro A, Coppedè F. Exposure to Metals, Pesticides, and Air Pollutants: Focus on Resulting DNA Methylation Changes in Neurodegenerative Diseases. Biomolecules 2024; 14:1366. [PMID: 39595543 PMCID: PMC11591912 DOI: 10.3390/biom14111366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Individuals affected by neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), are dramatically increasing worldwide. Thus, several efforts are being made to develop strategies for stopping or slowing the spread of these illnesses. Although causative genetic variants linked to the onset of these diseases are known, they can explain only a small portion of cases. The etiopathology underlying the neurodegenerative process in most of the patients is likely due to the interplay between predisposing genetic variants and environmental factors. Epigenetic mechanisms, including DNA methylation, are central candidates in translating the effects of environmental factors in genome modulation, and they play a critical role in the etiology of AD, PD, and ALS. Among the main environmental exposures that have been linked to an increased risk for these diseases, accumulating evidence points to the role of heavy metals, pesticides, and air pollutants. These compounds could trigger neurodegeneration through different mechanisms, mainly neuroinflammation and the induction of oxidative stress. However, increasing evidence suggests that they are also capable of inducing epigenetic alterations in neurons. In this article, we review the available literature linking exposure to metals, pesticides, and air pollutants to DNA methylation changes relevant to neurodegeneration.
Collapse
Affiliation(s)
- Andrea Stoccoro
- Laboratory of Medical Genetics, Department of Translational Research and of New Surgical and Medical Technologies, Medical School, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
| | - Fabio Coppedè
- Laboratory of Medical Genetics, Department of Translational Research and of New Surgical and Medical Technologies, Medical School, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- Interdepartmental Research Center of Biology and Pathology of Aging, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
42
|
Weishaupt AK, Ruecker L, Meiners T, Schwerdtle T, Silva Avila D, Aschner M, Bornhorst J. Copper-mediated neurotoxicity and genetic vulnerability in the background of neurodegenerative diseases in C. elegans. Toxicol Sci 2024; 201:254-262. [PMID: 39067045 PMCID: PMC11424883 DOI: 10.1093/toxsci/kfae092] [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] [Indexed: 07/30/2024] Open
Abstract
The mechanisms associated with neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), have yet to be fully characterized, and genetic as well as environmental factors in their disease etiology are underappreciated. Although mutations in genes such as PARKIN and LRRK2 have been linked to PD, the idiopathic component of the disease suggests a contribution of environmental risk factors, including metals, such as copper (Cu). Cu overexposure has been reported to cause oxidative stress and neurotoxicity, but its role in neurodegenerative diseases is rarely studied. Using Caenorhabditis elegans (C. elegans) as a model organism for neurotoxicity, we assessed the effects of Cu oversupply in AD and PD models. Our findings reveal that although copper treatment did not induce neurodegeneration in wild-type worms or the AD model, it significantly exacerbated neurodegeneration in the PD-associated mutants PARKIN and LRRK2. These results suggest that genetic predisposition for PD enhances the sensitivity to copper toxicity, highlighting the multifactorial nature of neurodegenerative diseases. Furthermore, our study provides insight into the mechanisms underlying Cu-induced neurotoxicity in PD models, including disruptions in dopamine levels, altered dopamine-dependent behavior and degraded dopaminergic neurons. Overall, our novel findings contribute to a better understanding of the complex interactions between genetic susceptibility, environmental factors, and neurodegenerative disease pathogenesis, emphasizing the importance of a tightly regulated Cu homeostasis in the etiology of PD. Copper oversupply exacerbated neurodegeneration in Caenorhabditis elegans models of Parkinson's disease, highlighting the genetic susceptibility and emphasizing the crucial role of tightly regulated copper homeostasis in Parkinson's disease pathogenesis.
Collapse
Affiliation(s)
- Ann-Kathrin Weishaupt
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
| | - Lysann Ruecker
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
| | - Torben Meiners
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
| | - Tanja Schwerdtle
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
- German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Daiana Silva Avila
- Laboratory of Toxicology and Biochemistry in Caenorhabditis elegans, Universidade Federal do Pampa, 97501-970 Uruguaiana, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Julia Bornhorst
- Food Chemistry with Focus on Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, 42119 Wuppertal, Germany
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Potsdam, Jena, Wuppertal, Germany
| |
Collapse
|
43
|
Zhu L, Du Y. A promising new approach to cancer therapy: Manipulate ferroptosis by hijacking endogenous iron. Int J Pharm 2024; 662:124517. [PMID: 39084581 DOI: 10.1016/j.ijpharm.2024.124517] [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: 04/08/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Ferroptosis, a form of regulated cell death characterized by iron-dependent phospholipid peroxidation, has emerged as a focal point in the field of cancer therapy. Compared with other cell death modes such as apoptosis and necrosis, ferroptosis exhibits many distinct characteristics in the molecular mechanisms and cell morphology, offering a promising avenue for combating cancers that are resistant to conventional therapeutic modalities. In light of the serious side effects associated with current Fenton-modulating ferroptosis therapies utilizing exogenous iron-based inorganic nanomaterials, hijacking endogenous iron could serve as an effective alternative strategy to trigger ferroptosis through targeting cellular iron regulatory mechanisms. A better understanding of the underlying iron regulatory mechanism in the process of ferroptosis has shed light on the current findings of endogenous ferroptosis-based nanomedicine strategies for cancer therapy. Here in this review article, we provide a comprehensive discussion on the regulatory network of iron metabolism and its pivotal role in ferroptosis, and present recent updates on the application of nanoparticles endowed with the ability to hijack endogenous iron for ferroptosis. We envision that the insights in the study may expedite the development and translation of endogenous ferroptosis-based nanomedicines for effective cancer treatment.
Collapse
Affiliation(s)
- Luwen Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321299, China.
| |
Collapse
|
44
|
Kale MB, Bhondge HM, Wankhede NL, Shende PV, Thanekaer RP, Aglawe MM, Rahangdale SR, Taksande BG, Pandit SB, Upaganlawar AB, Umekar MJ, Kopalli SR, Koppula S. Navigating the intersection: Diabetes and Alzheimer's intertwined relationship. Ageing Res Rev 2024; 100:102415. [PMID: 39002642 DOI: 10.1016/j.arr.2024.102415] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) and Diabetes mellitus (DM) exhibit comparable pathophysiological pathways. Genetic abnormalities in APP, PS-1, and PS-2 are linked to AD, with diagnostic aid from CSF and blood biomarkers. Insulin dysfunction, termed "type 3 diabetes mellitus" in AD, involves altered insulin signalling and neuronal shrinkage. Insulin influences beta-amyloid metabolism, exacerbating neurotoxicity in AD and amyloid production in DM. Both disorders display impaired glucose transporter expression, hastening cognitive decline. Mitochondrial dysfunction and Toll-like receptor 4-mediated inflammation worsen neurodegeneration in both diseases. ApoE4 raises disease risk, especially when coupled with dyslipidemia common in DM. Targeting shared pathways like insulin-degrading enzyme activation and HSP60 holds promise for therapeutic intervention. Recognizing these interconnected mechanisms underscores the imperative for developing tailored treatments addressing the overlapping pathophysiology of AD and DM, offering potential avenues for more effective management of both conditions.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | | | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Rushikesh P Thanekaer
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sunil B Pandit
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
45
|
Dave KM, Venna VR, Rao KS, Stolz DB, Brady B, Quaicoe VA, Maniskas ME, Hildebrand EE, Green D, Chen M, Milosevic J, Zheng SY, Shiva SS, McCullough LD, S Manickam D. Mitochondria-containing extracellular vesicles from mouse vs. human brain endothelial cells for ischemic stroke therapy. J Control Release 2024; 373:803-822. [PMID: 39084466 DOI: 10.1016/j.jconrel.2024.07.065] [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: 02/02/2024] [Revised: 07/10/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Ischemic stroke-induced mitochondrial dysfunction in the blood-brain barrier-forming brain endothelial cells (BECs) results in long-term neurological dysfunction post-stroke. We previously reported data from a pilot study where intravenous administration of human BEC (hBEC)-derived mitochondria-containing extracellular vesicles (EVs) showed a potential efficacy signal in a mouse middle cerebral artery occlusion (MCAo) model of stroke. We hypothesized that EVs harvested from donor species homologous to the recipient species (e.g., mouse) may improve therapeutic efficacy, and therefore, use of mouse BEC (mBEC)-derived EVs may improve post-stroke outcomes in MCAo mice. We investigated potential differences in the mitochondria transfer of EVs derived from the same species as the recipient cell (mBEC-EVs and recipient mBECs or hBECs-EVs and recipient hBECs) vs. cross-species EVs and recipient cells (mBEC-EVs and recipient hBECs or vice versa). Our results showed that while both hBEC- and mBEC-EVs transferred EV mitochondria, mBEC-EVs outperformed hBEC-EVs in increasing ATP levels and improved recipient mBEC mitochondrial function via increasing oxygen consumption rates. mBEC-EVs significantly reduced brain infarct volume and neurological deficit scores compared to vehicle-injected MCAo mice. The superior therapeutic efficacy of mBEC-EVs in MCAo mice support the continued use of mBEC-EVs to optimize the therapeutic potential of mitochondria-containing EVs in preclinical mouse models.
Collapse
Affiliation(s)
- Kandarp M Dave
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States of America
| | - Venugopal R Venna
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States of America
| | - Krithika S Rao
- Pittsburgh Heart Lung Blood Vascular Institute, University of Pittsburgh Medical School, Pittsburgh, PA, United States of America
| | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States of America
| | - Bodhi Brady
- Human Biology, South Dakota State University, Brookings, SD, United States of America
| | - Victoria A Quaicoe
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States of America
| | - Michael E Maniskas
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States of America
| | - Ella E Hildebrand
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States of America; Department of Psychology, Westminster College, New Wilmington, PA, United States of America
| | - Dawson Green
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States of America
| | - Mingxi Chen
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Jadranka Milosevic
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America; Captis Diagnostics Inc, Pittsburgh, PA, United States of America
| | - Si-Yang Zheng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Sruti S Shiva
- Pittsburgh Heart Lung Blood Vascular Institute, University of Pittsburgh Medical School, Pittsburgh, PA, United States of America; Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical School, Pittsburgh, PA, United States of America
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States of America
| | - Devika S Manickam
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States of America.
| |
Collapse
|
46
|
Mokhtar HI, Abd El-Fadeal NM, El-Sayed RM, Hegazy A, El-Kherbetawy MK, Hamad AG, ElSayed MH, Zaitone SA. Computational Analysis and Experimental Data Exploring the Role of Hesperetin in Ameliorating ADHD and SIRT1/Nrf2/Keap1/OH-1 Signaling. Int J Mol Sci 2024; 25:9284. [PMID: 39273230 PMCID: PMC11395673 DOI: 10.3390/ijms25179284] [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: 06/11/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 09/15/2024] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) manifests as poor attention, hyperactivity, as well as impulsive behaviors. Hesperetin (HSP) is a citrus flavanone with strong antioxidant and anti-inflammatory activities. The present study aimed to test hesperetin efficacy in alleviating experimental ADHD in mice and its influence on hippocampal neuron integrity and sirtuin 1 (SIRT1) signaling. An in silico study was performed to test the related proteins. Groups of mice were assigned as control, ADHD model, ADHD/HSP (25 mg/kg), and ADHD/HSP (50 mg/kg). ADHD was induced by feeding with monosodium glutamate (0.4 g/kg, for 8 weeks) and assessed by measuring the motor and attentive behaviors (open filed test, Y-maze test, and marble burying test), histopathological examination of the whole brain tissues, and estimation of inflammatory markers. The in-silico results indicated the putative effects of hesperetin on ADHD by allowing the integration and analysis of large-scale genomic, transcriptomic, and proteomic data. The in vivo results showed that ADHD model mice displayed motor hyperactivity and poor attention in the behavioral tasks and shrank neurons at various hippocampal regions. Further, there was a decline in the mRNA expression and protein levels for SIRT1, the erythroid 2-related factor-2 (Nrf2), kelch like ECH associated protein 1 (Keap1) and hemeoxygenase-1 (OH-1) proteins. Treatment with HSP normalized the motor and attentive behaviors, prevented hippocampal neuron shrinkage, and upregulated SIRT1/Nrf2/Keap1/OH-1 proteins. Taken together, HSP mainly acts by its antioxidant potential. However, therapeutic interventions with hesperetin or a hesperetin-rich diet can be suggested as a complementary treatment in ADHD patients but cannot be suggested as an ADHD treatment per se as it is a heterogeneous and complex disease.
Collapse
Affiliation(s)
- Hatem I. Mokhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University-Kantara Branch, Ismailia 41636, Egypt
| | - Noha M. Abd El-Fadeal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Rehab M. El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University—Arish Branch, Arish, 45511, Egypt
| | - Ann Hegazy
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | | | - Ahmed G. Hamad
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed H. ElSayed
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo 11757, Egypt
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| |
Collapse
|
47
|
Vannelli A, Mariano V, Bagni C, Kanellopoulos AK. Activation of the 5-HT1A Receptor by Eltoprazine Restores Mitochondrial and Motor Deficits in a Drosophila Model of Fragile X Syndrome. Int J Mol Sci 2024; 25:8787. [PMID: 39201473 PMCID: PMC11354613 DOI: 10.3390/ijms25168787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Neurons rely on mitochondrial energy metabolism for essential functions like neurogenesis, neurotransmission, and synaptic plasticity. Mitochondrial dysfunctions are associated with neurodevelopmental disorders including Fragile X syndrome (FXS), the most common cause of inherited intellectual disability, which also presents with motor skill deficits. However, the precise role of mitochondria in the pathophysiology of FXS remains largely unknown. Notably, previous studies have linked the serotonergic system and mitochondrial activity to FXS. Our study investigates the potential therapeutic role of serotonin receptor 1A (5-HT1A) in FXS. Using the Drosophila model of FXS, we demonstrated that treatment with eltoprazine, a 5-HT1A agonist, can ameliorate synaptic transmission, correct mitochondrial deficits, and ultimately improve motor behavior. While these findings suggest that the 5-HT1A-mitochondrial axis may be a promising therapeutic target, further investigation is needed in the context of FXS.
Collapse
Affiliation(s)
- Anna Vannelli
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | | |
Collapse
|
48
|
Tenchov R, Sasso JM, Zhou QA. Polyglutamine (PolyQ) Diseases: Navigating the Landscape of Neurodegeneration. ACS Chem Neurosci 2024; 15:2665-2694. [PMID: 38996083 PMCID: PMC11311141 DOI: 10.1021/acschemneuro.4c00184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/02/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
Polyglutamine (polyQ) diseases are a group of inherited neurodegenerative disorders caused by expanded cytosine-adenine-guanine (CAG) repeats encoding proteins with abnormally expanded polyglutamine tract. A total of nine polyQ disorders have been identified, including Huntington's disease, six spinocerebellar ataxias, dentatorubral pallidoluysian atrophy (DRPLA), and spinal and bulbar muscular atrophy (SBMA). The diseases of this class are each considered rare, yet polyQ diseases constitute the largest group of monogenic neurodegenerative disorders. While each subtype of polyQ diseases has its own causative gene, certain pathologic molecular attributes have been implicated in virtually all of the polyQ diseases, including protein aggregation, proteolytic cleavage, neuronal dysfunction, transcription dysregulation, autophagy impairment, and mitochondrial dysfunction. Although animal models of polyQ disease are available helping to understand their pathogenesis and access disease-modifying therapies, there is neither a cure nor prevention for these diseases, with only symptomatic treatments available. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in the class of polyQ diseases. We examine the publication landscape in the area in effort to provide insights into current knowledge advances and developments. We review the most discussed concepts and assess the strategies to combat these diseases. Finally, we inspect clinical applications of products against polyQ diseases with their development pipelines. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding the class of polyQ diseases, to outline challenges, and evaluate growth opportunities to further efforts in combating the diseases.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American
Chemical Society, Columbus, Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American
Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
49
|
Sayehmiri F, Motamedi F, Batool Z, Naderi N, Shaerzadeh F, Zoghi A, Rezaei O, Khodagholi F, Pourbadie HG. Mitochondrial plasticity and synaptic plasticity crosstalk; in health and Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14897. [PMID: 39097920 PMCID: PMC11298206 DOI: 10.1111/cns.14897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024] Open
Abstract
Synaptic plasticity is believed to underlie the cellular and molecular basis of memory formation. Mitochondria are one of the main organelles involved in metabolism and energy maintenance as plastic organelles that change morphologically and functionally in response to cellular needs and regulate synaptic function and plasticity through multiple mechanisms, including ATP generation, calcium homeostasis, and biogenesis. An increased neuronal activity enhances synaptic efficiency, during which mitochondria's spatial distribution and morphology change significantly. These organelles build up in the pre-and postsynaptic zones to produce ATP, which is necessary for several synaptic processes like neurotransmitter release and recycling. Mitochondria also regulate calcium homeostasis by buffering intracellular calcium, which ensures proper synaptic activity. Furthermore, mitochondria in the presynaptic terminal have distinct morphological properties compared to dendritic or postsynaptic mitochondria. This specialization enables precise control of synaptic activity and plasticity. Mitochondrial dysfunction has been linked to synaptic failure in many neurodegenerative disorders, like Alzheimer's disease (AD). In AD, malfunctioning mitochondria cause delays in synaptic vesicle release and recycling, ionic gradient imbalances, and mostly synaptic failure. This review emphasizes mitochondrial plasticity's contribution to synaptic function. It also explores the profound effect of mitochondrial malfunction on neurodegenerative disorders, focusing on AD, and provides an overview of how they sustain cellular health under normal conditions and how their malfunction contributes to neurodegenerative diseases, highlighting their potential as a therapeutic target for such conditions.
Collapse
Affiliation(s)
- Fatemeh Sayehmiri
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
- Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Nima Naderi
- Department of Pharmacology and Toxicology, Faculty of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | | | - Anahita Zoghi
- Department of Neurology, Loghman Hakim HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Omidvar Rezaei
- Skull Base Research CenterLoghman Hakim Hospital, Shahid Beheshti University of Medical SciencesTehranIran
| | - Fariba Khodagholi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
50
|
Calabrese V, Osakabe N, Siracusa R, Modafferi S, Di Paola R, Cuzzocrea S, Jacob UM, Fritsch T, Abdelhameed AS, Rashan L, Wenzel U, Franceschi C, Calabrese EJ. Transgenerational hormesis in healthy aging and antiaging medicine from bench to clinics: Role of food components. Mech Ageing Dev 2024; 220:111960. [PMID: 38971236 DOI: 10.1016/j.mad.2024.111960] [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: 04/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
Neurodegenerative diseases have multifactorial pathogenesis, mainly involving neuroinflammatory processes. Finding drugs able to treat these diseases, expecially because for most of these diseases there are no effective drugs, and the current drugs cause undesired side effects, represent a crucial point. Most in vivo and in vitro studies have been concentrated on various aspects related to neurons (e.g. neuroprotection), however, there has not been focus on the prevention of early stages involving glial cell activation and neuroinflammation. Recently, it has been demonstrated that nutritional phytochemicals including polyphenols, the main active constituents of the Mediterranean diet, maintain redox balance and neuroprotection through the activation of hormetic vitagene pathway. Recent lipidomics data from our laboratory indicate mushrooms as strong nutritional neuronutrients with strongly activity against neuroinflammation in Meniere' diseaseas, a model of cochleovestibular neural degeneration, as well as in animal model of traumatic brain injury, or rotenone induced parkinson's disease. Moreover, Hidrox®, an aqueous extract of olive containing hydroxytyrosol, and Boswellia, acting as Nrf2 activators, promote resilience by enhancing the redox potential, and thus, regulate through hormetic mechanisms, cellular stress response mechanisms., Thus, modulation of cellular stress pathways, in particular vitagenes system, may be an innovative approach for therapeutic intervention in neurodegenerative disorders.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Naomi Osakabe
- Department of Bioscience and Engineering, Shibaura Institute Technology, Tokyo, Japan.
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | - Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Messina 98168, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | | | | | - Ali S Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Luay Rashan
- Biodiversity Unit, Dhofar University, Salalah, Oman
| | - Uwe Wenzel
- Institut für Ernährungswissenschaft, Justus Liebig Universitat Giessen, Germany
| | | | - Edward J Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|